1
|
Kharrat A, Diambomba Y, Jain A. Corticosteroid use in neonatal hypotension: A survey of Canadian neonatologists. Pediatr Neonatol 2024; 65:451-456. [PMID: 38388227 DOI: 10.1016/j.pedneo.2023.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 02/24/2024] Open
Abstract
OBJECTIVE To assess prescribing practices and perspectives regarding the use of corticosteroids in the management of neonatal hypotension. METHODS Cross-sectional questionnaire-based electronic survey of neonatologists (n = 206) practicing at tertiary neonatal intensive care units across 30 academic centres in Canada. RESULTS The overall response rate was 33% (72/206), with a completion rate was 94%. Most (48/72, 64%) worked in a unit that covered both inborn and outborn infants, and 53% (37/70) worked in units with >100 very low birth weight infants admitted annually. Among the 72 respondents, 39% use a loading dose, of whom most (57%) use 2 mg/kg. Dosing ranges were variable, most using either 0.5 mg/kg or 1 mg/kg, q6h. Among the 56% (40/72) of neonatologists who reported measuring cortisol before initiation of hydrocortisone, cut-offs for initiation of hydrocortisone varied from <100 to <500 nmol/L, most of whom (48%) used <100 nmol/L. Of 71 respondents, 92% (65) indicated that a randomized control trial examining the use of corticosteroids in neonatal hypotension is needed, of whom 52% (37) indicated that the intervention group should receiving hydrocortisone after one vasopressor/inotrope. CONCLUSIONS This survey provides insight into the prescribing practices of tertiary neonatologists with regards to the use of corticosteroids in neonatal hypotension. While corticosteroids are frequently prescribed, there is variability in the indication, dosing, and duration of corticosteroid use. The findings from this survey can be used to inform further research, including a clinical trial, regarding the practice in the management of neonatal hypotension.
Collapse
Affiliation(s)
- Ashraf Kharrat
- Department of Paediatrics, Mount Sinai Hospital, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada.
| | - Yenge Diambomba
- Department of Paediatrics, Mount Sinai Hospital, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| | - Amish Jain
- Department of Paediatrics, Mount Sinai Hospital, Toronto, ON, Canada; Department of Paediatrics, University of Toronto, Toronto, ON, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| |
Collapse
|
2
|
Zou H, Yu H, Huang Y, Guo Y, Ye M, Hou L. Chronic exposure to gestodene impairs reproductive system in adult female zebrafish (Daniarerio). CHEMOSPHERE 2024; 355:141876. [PMID: 38570043 DOI: 10.1016/j.chemosphere.2024.141876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/11/2024] [Accepted: 03/30/2024] [Indexed: 04/05/2024]
Abstract
Gestodene (GES) is widely used in human therapy and animal husbandry and is frequently detected in aquatic environments. Although GES adversely affects aquatic organisms at trace levels, its effects on the reproductive biology of fish remain inconclusive. In this study, female zebrafish (Danio rerio) were exposed to environmentally relevant levels of GES for the evaluation of the effects of GES on the reproductive system by using endpoints including gene expression, plasma steroid concentrations, histological and morphological analyses, copulatory behavior, and reproductive output. Adult female zebrafish exposed to environmentally relevant concentrations of GES (4.0, 40.2, and 372.7 ng/L) for 60 d demonstrated stagnant ovarian oocyte development, evidenced by an increase in the percentage of perinuclear and atretic oocytes and a decrease in the percentage of late vitellogenic oocytes. GES-exposed females were less attractive to males and had lower copulatory intimacy than females in control. Consequently, spawning (44.3-49.2 %) and egg fertilization rates (27.9-32.0 %) were decreased. The decreased survival of fertilized eggs and hatching rates were accompanied by increased malformations. These negative effects were associated with abnormal transcriptional levels of gonadal steroid hormones, which were regulated by genes (Hsd17β3, Hsd11β2, Hsd20β, Cyp19a1a, and Cyp11b). Overall, our findings suggest that GES impairs the reproductive system of zebrafish, which may threaten population stability.
Collapse
Affiliation(s)
- Hong Zou
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - HongJun Yu
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - YunYi Huang
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - YanFang Guo
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - MeiXin Ye
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China
| | - LiPing Hou
- School of Life Sciences, Guangzhou University, Guangzhou 510655, China; Key Laboratory of Conservation and Application in Biodiversity of South China, School of Life Sciences, Guangzhou University, Guangzhou, Guangdong, 510006, China.
| |
Collapse
|
3
|
Nethathe GD, Lipman J, Anderson R, Fuller PJ, Feldman C. Glucocorticoids with or without fludrocortisone in septic shock: a narrative review from a biochemical and molecular perspective. Br J Anaesth 2024; 132:53-65. [PMID: 38030548 PMCID: PMC10797514 DOI: 10.1016/j.bja.2023.10.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Two randomised controlled trials have reported a reduction in mortality when adjunctive hydrocortisone is administered in combination with fludrocortisone compared with placebo in septic shock. A third trial did not support this finding when hydrocortisone administered in combination with fludrocortisone was compared with hydrocortisone alone. The underlying mechanisms for this mortality benefit remain poorly understood. We review the clinical implications and potential mechanisms derived from laboratory and clinical data underlying the beneficial role of adjunctive fludrocortisone with hydrocortisone supplementation in septic shock. Factors including distinct biological effects of glucocorticoids and mineralocorticoids, tissue-specific and mineralocorticoid receptor-independent effects of mineralocorticoids, and differences in downstream signalling pathways between mineralocorticoid and glucocorticoid binding at the mineralocorticoid receptor could contribute to this interaction. Furthermore, pharmacokinetic and pharmacodynamic disparities exist between aldosterone and its synthetic counterpart fludrocortisone, potentially influencing their effects. Pending publication of well-designed, randomised controlled trials, a molecular perspective offers valuable insights and guidance to help inform clinical strategies.
Collapse
Affiliation(s)
- Gladness D Nethathe
- School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa; Department of Anaesthesia and Perioperative Medicine, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia; Academy of Critical Care, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia.
| | - Jeffrey Lipman
- Academy of Critical Care, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia; Jamieson Trauma Institute and Intensive Care Services, Royal Brisbane and Women's Hospital, Butterfield Street, Herston, Brisbane, 4029, QLD, Australia; Nimes University Hospital, University of Montpellier, Nimes, France
| | - Ronald Anderson
- Department of Immunology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Peter J Fuller
- Endocrinology Unit, Monash Health, Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research, Melbourne, VIC, Australia
| | - Charles Feldman
- School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
4
|
Ben-Eltriki M, Shankar G, Tomlinson Guns ES, Deb S. Pharmacokinetics and pharmacodynamics of Rh2 and aPPD ginsenosides in prostate cancer: a drug interaction perspective. Cancer Chemother Pharmacol 2023; 92:419-437. [PMID: 37709921 DOI: 10.1007/s00280-023-04583-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023]
Abstract
Ginsenoside Rh2 and its aglycon (aPPD) are one of the major metabolites from Panax ginseng. Preclinical studies suggest that Rh2 and aPPD have antitumor effects in prostate cancer (PCa). Our aims in this review are (1) to describe the pharmacokinetic (PK) properties of Rh2 and aPPD ginsenosides; 2) to provide an overview of the preclinical findings on the use of Rh2 and aPPD in the treatment of PCa; and (3) to highlight the mechanisms of its PK and pharmacodynamic (PD) drug interactions. Increasing evidence points to the potential efficacy of Rh2 or aPPD for PCa treatment. Based on the laboratory studies, Rh2 or aPPD combinations revealed an additive or synergistic interaction or enhanced sensitivity of anticancer drugs toward PCa. This review reveals that enhanced anticancer activities were demonstrated in preclinical studies through interactions of Rh2 and/or aPPD with the proteins related to PK (e.g., cytochrome P450 enzymes, transporters) or PD of the other anticancer drugs or PCa signaling pathways. In conclusion, combining Rh2 or aPPD with anti-prostate cancer drugs leads to PK or PD interactions which could facilitate either therapeutically beneficial or toxic effects.
Collapse
Affiliation(s)
- Mohamed Ben-Eltriki
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada.
- Cochrane Hypertension Review Group, Therapeutic Initiative, University of British Columbia, Vancouver, BC, Canada.
- Community Pharmacist, Vancouver Area, BC, Canada.
- Department of Pharmacology and Therapeutics, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Gehana Shankar
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Emma S Tomlinson Guns
- The Vancouver Prostate Centre at Vancouver General Hospital, 2660 Oak Street, Vancouver, BC, V6H 3Z6, Canada
| | - Subrata Deb
- Department of Pharmaceutical Sciences, College of Pharmacy, Larkin University, Miami, FL, 33169, USA.
| |
Collapse
|
5
|
Liang J, Liu QS, Ren Z, Min K, Yang X, Hao F, Zhang Q, Liu Q, Zhou Q, Jiang G. Studying paraben-induced estrogen receptor- and steroid hormone-related endocrine disruption effects via multi-level approaches. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 869:161793. [PMID: 36702264 DOI: 10.1016/j.scitotenv.2023.161793] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/19/2023] [Accepted: 01/19/2023] [Indexed: 06/18/2023]
Abstract
Increasing concerns have been raised on the health risks of parabens in the regard of their widespread applications and potential endocrine disrupting activities. In this study, four typical parabens, including methyl paraben (MeP), ethyl paraben (EtP), propyl paraben (PrP), and butyl paraben (BuP) were systematically investigated for their estrogen receptor- and steroid hormone-related endocrine disruptions using multi-level approaches. Paraben exposure promoted the proliferation of MCF-7 cells, increased the luciferase activity in MVLN cells, and induced the vitellogenin (vtg) expression in zebrafish larvae, showing the typical estrogenic effects. The in vitro protein assays further revealed that PrP and BuP could bind with two isoforms of estrogen receptors (ERs). The estrogenic activities of parabens were predicted to be positively correlated with their chemical structure complexity by using molecular docking analysis. Furthermore, the synthesis and secretion of estradiol (E2) and testosterone (T) were significantly disturbed in H295R cells and zebrafish larvae, which could be regulated by paraben-induced transcriptional disturbance in both in vitro steroidogenesis and in vivo hypothalamic-pituitary-gonadal (HPG) axis. Parabens could disturb the endocrine system by activating the ERs and disrupting the steroid hormone synthesis and secretion, suggesting their potential deleterious risks to the environment and human health.
Collapse
Affiliation(s)
- Jiefeng Liang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, PR China; Shandong Key Laboratory of Environmental Processes and Health, School of Environmental Science and Engineering, Shandong University, Qingdao 266237, PR China
| | - Qian S Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China.
| | - Zhihua Ren
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Ke Min
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Xiaoxi Yang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Fang Hao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; Institute of Analytical Food Safety, School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Qing Zhang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China
| | - Qian Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Qunfang Zhou
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| | - Guibin Jiang
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, PR China; College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, PR China
| |
Collapse
|
6
|
Cory MJ, Durand P, Sillero R, Morin L, Savani R, Chalak L, Angelis D. Vein of Galen aneurysmal malformation: rationalizing medical management of neonatal heart failure. Pediatr Res 2023; 93:39-48. [PMID: 35422084 DOI: 10.1038/s41390-022-02064-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/21/2022] [Accepted: 03/26/2022] [Indexed: 01/28/2023]
Abstract
Neonates who present in high output heart failure secondary to vein of Galen aneurysmal malformation can be difficult to manage medically due to the complex physiology that results from the large shunt through the malformation. Though the cardiac function is often normal, right ventricular dilation, severe pulmonary hypertension, and systemic steal can result in inadequate organ perfusion and shock. This report recommends medical management for stabilization of neonates prior to definitive management with endovascular embolization. IMPACT: Vein of Galen aneurysmal malformation (VGAM) is a rare intracranial arteriovenous malformation, which can present in the neonatal period with high output heart failure. Heart failure secondary to VGAM is often difficult to manage and is associated with high mortality and morbidity. Despite optimal medical management, many patients require urgent endovascular embolization for stabilization of their heart failure. This report offers discrete recommendations that can be used by clinicians as guidelines for the medical management of heart failure in newborns with VGAM.
Collapse
Affiliation(s)
- Melinda J Cory
- Division of Cardiology, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Phillippe Durand
- Division of Pediatrics and Neonatal Intensive Care, Paris-Saclay University Hospitals, Bicêtre Medical Centre, Assistance Publique-Hospitaux de Paris, Paris, France
| | - Rafael Sillero
- Division of Neurosurgery, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Luc Morin
- Division of Pediatrics and Neonatal Intensive Care, Paris-Saclay University Hospitals, Bicêtre Medical Centre, Assistance Publique-Hospitaux de Paris, Paris, France
| | - Rashmin Savani
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lina Chalak
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dimitrios Angelis
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
7
|
Almeida ZL, Brito RMM. Amyloid Disassembly: What Can We Learn from Chaperones? Biomedicines 2022; 10:3276. [PMID: 36552032 PMCID: PMC9776232 DOI: 10.3390/biomedicines10123276] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/14/2022] [Accepted: 09/26/2022] [Indexed: 12/23/2022] Open
Abstract
Protein aggregation and subsequent accumulation of insoluble amyloid fibrils with cross-β structure is an intrinsic characteristic of amyloid diseases, i.e., amyloidoses. Amyloid formation involves a series of on-pathway and off-pathway protein aggregation events, leading to mature insoluble fibrils that eventually accumulate in multiple tissues. In this cascade of events, soluble oligomeric species are formed, which are among the most cytotoxic molecular entities along the amyloid cascade. The direct or indirect action of these amyloid soluble oligomers and amyloid protofibrils and fibrils in several tissues and organs lead to cell death in some cases and organ disfunction in general. There are dozens of different proteins and peptides causing multiple amyloid pathologies, chief among them Alzheimer's, Parkinson's, Huntington's, and several other neurodegenerative diseases. Amyloid fibril disassembly is among the disease-modifying therapeutic strategies being pursued to overcome amyloid pathologies. The clearance of preformed amyloids and consequently the arresting of the progression of organ deterioration may increase patient survival and quality of life. In this review, we compiled from the literature many examples of chemical and biochemical agents able to disaggregate preformed amyloids, which have been classified as molecular chaperones, chemical chaperones, and pharmacological chaperones. We focused on their mode of action, chemical structure, interactions with the fibrillar structures, morphology and toxicity of the disaggregation products, and the potential use of disaggregation agents as a treatment option in amyloidosis.
Collapse
Affiliation(s)
| | - Rui M. M. Brito
- Chemistry Department and Coimbra Chemistry Centre—Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
8
|
Zhu T, Zhou Q, Yang Z, Zhang Y, Luo J, Zhang X, Shen Y, Jiao L, Tocher DR, Jin M. Dietary cholesterol promotes growth and ecdysone signalling pathway by modulating cholesterol transport in swimming crabs (Portunus trituberculatus). ANIMAL NUTRITION 2022; 10:249-260. [PMID: 35785252 PMCID: PMC9234081 DOI: 10.1016/j.aninu.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/12/2022] [Accepted: 05/09/2022] [Indexed: 11/29/2022]
Abstract
Cholesterol, as an indispensable nutrient, regulates molting and growth in crustacean. As crustaceans are unable to biosynthesize cholesterol de novo, it is central to understand how dietary cholesterol affects molting in crustaceans. An 8-week feeding trial was conducted to evaluate the effects of dietary cholesterol level (0.12%, 0.43%, 0.79%, 1.00%, 1.30% and 2.50%) on growth, cholesterol metabolism and expression of genes related to lipid and ecdysone metabolism in female swimming crabs (Portunus trituberculatus). A total of 192 crabs (1.41 ± 0.05 g) were randomly distributed into 192 aquaria. Each treatment had 4 replicates with each replicate containing 8 crabs. Crabs fed the 1.00% cholesterol diet showed best growth performance, and thus based on percent weight gain, the optimal dietary cholesterol requirement was calculated at 1.01%. Tissue cholesterol concentrations were positively correlated with dietary cholesterol level. The contents of functional fatty acids in hepatopancreas significantly increased as dietary cholesterol increased from 0.12% to 2.50% (P < 0.05). The expression levels of genes related to lipogenesis pathway, lipid catabolism and fatty acid oxidation were significantly down-regulated with increased dietary cholesterol level (P < 0.05). The highest expression levels of cholesterol transport genes, low-density lipoprotein receptor (ldlr) and low-density lipoprotein receptor-related protein 2 (lrp2) occurred in crabs fed the 1.30% cholesterol diet. Moreover, hormones related to molting such as crustacean hyperglycemic hormone (CHH), methyl farnesoate (MF), molt-inhibiting hormone (MIH), and ecdysone in hemolymph were significantly influenced by dietary cholesterol level (P < 0.05). The highest expression levels of ecdysone receptor (ecr) and chitinase 1 (chi1) in eyestalk and hepatopancreas were found in crabs fed the diet containing 1.00% cholesterol (P < 0.05). In conclusion, the optimal dietary level was beneficial to functional fatty acid accumulation, regulated lipid metabolism, promoted the ecdysone signalling pathway by improving the cholesterol transport, and improved the molting rate and growth of swimming crabs.
Collapse
Affiliation(s)
- Tingting Zhu
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
| | - Qicun Zhou
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
- Corresponding authors.
| | - Zheng Yang
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
| | - Yingying Zhang
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
| | - Jiaxiang Luo
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
| | - Xiangsheng Zhang
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
| | - Yuedong Shen
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
| | - Lefei Jiao
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
| | - Douglas R. Tocher
- Guangdong Provincial Key Laboratory of Marine Biotechnology, Institute of Marine Sciences, Shantou University, 515063 Shantou, China
| | - Min Jin
- Laboratory of Fish and Shellfish Nutrition, School of Marine Sciences, Ningbo University, 315211 Ningbo, China
- Corresponding authors.
| |
Collapse
|
9
|
Perusquía M. Androgens and Non-Genomic vascular responses in hypertension. Biochem Pharmacol 2022; 203:115200. [PMID: 35926652 DOI: 10.1016/j.bcp.2022.115200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/02/2022]
Abstract
Arterial hypertension is a global public health concern. In the last few years, the interest in androgen deficiency has been growing, and the association between androgens and high blood pressure (BP) is still controversial. One purpose of this review was to summarize the available findings in order to clarify whether male sex steroid hormones have beneficial or harmful effect on BP. The second purpose was to enhance the recognition of the acute non-genomic sex-independent vasorelaxing effect of androgens. Remarkably, BP variation is expected to be a consequence of the androgen-induced vasorelaxation which reduces systemic BP; hence the in vivo vasodepressor, hypotensive, and antihypertensive responses of androgens were also analyzed. This article reviews the current understanding of the physiological regulation of vascular smooth muscle contractility by androgens. Additionally, it summarizes older and more recent data on androgens, and some of the possible underlying mechanisms of relaxation, structural-functional differences in the androgen molecules, and their designing ability to induce vasorelaxation. The clinical relevance of these findings in terms of designing future therapeutics mainly the 5-reduced metabolite of testosterone, 5β-dihydrotestosterone, is also highlighted. Literature collected through a PubMed database search, as well as our experimental work, was used for the present review.
Collapse
Affiliation(s)
- Mercedes Perusquía
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, 04510 Ciudad de México, México.
| |
Collapse
|
10
|
Kamps NN, Banks R, Reeder RW, Berg RA, Newth CJ, Pollack MM, Meert KL, Carcillo JA, Mourani PM, Sorenson S, Varni JW, Cengiz P, Zimmerman JJ. The Association of Early Corticosteroid Therapy With Clinical and Health-Related Quality of Life Outcomes in Children With Septic Shock. Pediatr Crit Care Med 2022; 23:687-697. [PMID: 35695852 PMCID: PMC9444900 DOI: 10.1097/pcc.0000000000003009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVES Corticosteroids are commonly used in the treatment of pediatric septic shock without clear evidence of the potential benefits or risks. This study examined the association of early corticosteroid therapy with patient-centered clinically meaningful outcomes. DESIGN Subsequent cohort analysis of data derived from the prospective Life After Pediatric Sepsis Evaluation (LAPSE) investigation. Outcomes among patients receiving hydrocortisone or methylprednisolone on study day 0 or 1 were compared with those who did not use a propensity score-weighted analysis that controlled for age, sex, study site, and measures of first-day illness severity. SETTING Twelve academic PICUs in the United States. PATIENTS Children with community-acquired septic shock 1 month to 18 years old enrolled in LAPSE, 2013-2017. Exclusion criteria included a history of chronic corticosteroid administration. INTERVENTIONS None. MEASUREMENTS AND MAIN RESULTS Among children enrolled in LAPSE, 352 of 392 met analysis inclusion criteria, and 155 of 352 (44%) received early corticosteroid therapy. After weighting corticosteroid therapy administration propensity across potentially confounding baseline characteristics, differences in outcomes associated with treatment were not statistically significant (adjusted effect or odds ratio [95% CI]): vasoactive-inotropic support duration (-0.37 d [-1.47 to 0.72]; p = 0.503), short-term survival without new morbidity (1.37 [0.83-2.28]; p = 0.218), new morbidity among month-1 survivors (0.70 [0.39-1.23]; p = 0.218), and persistent severe deterioration of health-related quality of life or mortality at month 1 (0.70 [0.40-1.23]; p = 0.212). CONCLUSIONS This study examined the association of early corticosteroid therapy with mortality and morbidity among children encountering septic shock. After adjusting for variables with the potential to confound the relationship between early corticosteroid administration and clinically meaningful end points, there was no improvement in outcomes associated with this therapy. Results from this propensity analysis provide additional justification for equipoise regarding corticosteroid therapy for pediatric septic shock and ascertain the need for a well-designed clinical trial to examine benefit/risk for this intervention.
Collapse
Affiliation(s)
- Nicole N Kamps
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, American Family Children's Hospital, University of Wisconsin, Madison, WI
| | - Russell Banks
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Utah, Salt Lake City, UT
| | - Ron W Reeder
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Utah, Salt Lake City, UT
| | - Robert A Berg
- Department of Anesthesiology and Critical Care Medicine Children's Hospital of Philadelphia, Philadelphia, PA
| | - Christopher J Newth
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital Los Angeles, Los Angeles, CA
| | - Murray M Pollack
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Children's National Hospital, Washington, DC
| | - Kathleen L Meert
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Children's Hospital of Michigan, Detroit, MI
- Department of Pediatrics, Central Michigan University, Mt. Pleasant, MI
| | - Joseph A Carcillo
- Department of Critical Care Medicine, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA
| | - Peter M Mourani
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Colorado School of Medicine, Children's Hospital Colorado, Aurora, CO
| | - Samuel Sorenson
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, University of Utah, Salt Lake City, UT
| | - James W Varni
- Department of Pediatrics, Texas A&M University, College Station, TX
| | - Pelin Cengiz
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, American Family Children's Hospital, University of Wisconsin, Madison, WI
| | - Jerry J Zimmerman
- Division of Pediatric Critical Care Medicine, Department of Pediatrics, Seattle Children's Hospital, Seattle Children's Research Institute, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
11
|
Glomerular Mesangial Cell pH Homeostasis Mediates Mineralocorticoid Receptor-Induced Cell Proliferation. Biomedicines 2021; 9:biomedicines9091117. [PMID: 34572303 PMCID: PMC8468551 DOI: 10.3390/biomedicines9091117] [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: 07/14/2021] [Revised: 08/24/2021] [Accepted: 08/27/2021] [Indexed: 11/17/2022] Open
Abstract
Mineralocorticoids (e.g., aldosterone) support chronic inflammatory tissue damage, including glomerular mesangial injury leading to glomerulosclerosis. Furthermore, aldosterone leads to activation of the extracellular signal-regulated kinases (ERK1/2) in rat glomerular mesangial cells (GMC). Because ERK1/2 can affect cellular pH homeostasis via activation of Na+/H+-exchange (NHE) and the resulting cellular alkalinization may support proliferation, we tested the hypothesis that aldosterone affects pH homeostasis and thereby cell proliferation as well as collagen secretion also in primary rat GMC. Cytoplasmic pH and calcium were assessed by single-cell fluorescence ratio imaging, using the dyes BCECF or FURA2, respectively. Proliferation was determined by cell counting, thymidine incorporation and collagen secretion by collagenase-sensitive proline incorporation and ERK1/2-phosphorylation by Western blot. Nanomolar aldosterone induces a rapid cytosolic alkalinization which is prevented by NHE inhibition (10 µmol/L EIPA) and by blockade of the mineralocorticoid receptor (100 nmol/L spironolactone). pH changes were not affected by inhibition of HCO3- transporters and were not dependent on HCO3-. Aldosterone enhanced ERK1/2 phosphorylation and inhibition of ERK1/2-phosphorylation (10 µmol/L U0126) prevented aldosterone-induced alkalinization. Furthermore, aldosterone induced proliferation of GMC and collagen secretion, both of which were prevented by U0126 and EIPA. Cytosolic calcium was not involved in this aldosterone action. In conclusion, our data show that aldosterone can induce GMC proliferation via a MR and ERK1/2-mediated activation of NHE with subsequent cytosolic alkalinization. GMC proliferation leads to glomerular hypercellularity and dysfunction. This effect presents a possible mechanism contributing to mineralocorticoid receptor-induced pathogenesis of glomerular mesangial injury during chronic kidney disease.
Collapse
|
12
|
Joshua Cohen D, ElBaradie K, Boyan BD, Schwartz Z. Sex-specific effects of 17β-estradiol and dihydrotestosterone (DHT) on growth plate chondrocytes are dependent on both ERα and ERβ and require palmitoylation to translocate the receptors to the plasma membrane. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:159028. [PMID: 34416391 DOI: 10.1016/j.bbalip.2021.159028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/19/2021] [Accepted: 08/13/2021] [Indexed: 11/24/2022]
Abstract
Rat costochondral cartilage growth plate chondrocytes exhibit cell sex-specific responses to 17β-estradiol (E2), testosterone, and dihydrotestosterone (DHT). Mechanistically, E2 and DHT stimulate proliferation and extracellular matrix synthesis in chondrocytes from female and male rats, respectively, by signaling through protein kinase C (PKC) and phospholipase C (PLC). Estrogen receptors (ERα; ERβ) and androgen receptors (ARs) are present in both male and female cells, but it is not known whether they interact to elicit sex-specific signaling. We used specific agonists and antagonists of these receptors to examine the relative contributions of ERs and ARs in membrane-mediated E2 signaling in female chondrocytes and DHT signaling in male chondrocytes. PKC activity in female chondrocytes was stimulated by agonists of ERα and ERβ and required intact caveolae; PKC activity was inhibited by the E2 enantiomer and by an inhibitor of ERβ. Western blots of cell lysates co-immunoprecipitated for ERα suggested the formation of a complex containing both ERα and ERß with E2 treatment. DHT and DHT agonists activated PKC in male cells, while AR inhibition blocked the stimulatory effect of DHT on PKC. Inhibition of ERα and ERβ also blocked PKC activation by DHT. Western blots of whole-cell lysates, plasma membranes, and caveolae indicated the translocation of AR to the plasma membrane and specifically to caveolae with DHT treatment. These results suggest that E2 and DHT promote chondrocyte differentiation via the ability of ARs and ERs to form a complex. The results also indicate that intact caveolae and palmitoylation of the membrane receptor(s) or membrane receptor complex containing ERα and ERβ is required for E2 and DHT membrane-associated PKC activity in costochondral cartilage cells.
Collapse
Affiliation(s)
- D Joshua Cohen
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Khairat ElBaradie
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30033, USA
| | - Barbara D Boyan
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30033, USA.
| | - Zvi Schwartz
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA 23284, USA; Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
13
|
|
14
|
Cao W, Luo C, Lei M, Shen M, Ding W, Wang M, Song M, Ge J, Zhang Q. Development and Validation of a Dynamic Nomogram to Predict the Risk of Neonatal White Matter Damage. Front Hum Neurosci 2021; 14:584236. [PMID: 33708079 PMCID: PMC7940363 DOI: 10.3389/fnhum.2020.584236] [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: 07/16/2020] [Accepted: 12/31/2020] [Indexed: 12/23/2022] Open
Abstract
Purpose White matter damage (WMD) was defined as the appearance of rough and uneven echo enhancement in the white matter around the ventricle. The aim of this study was to develop and validate a risk prediction model for neonatal WMD. Materials and Methods We collected data for 1,733 infants hospitalized at the Department of Neonatology at The First Affiliated Hospital of Zhengzhou University from 2017 to 2020. Infants were randomly assigned to training (n = 1,216) or validation (n = 517) cohorts at a ratio of 7:3. Multivariate logistic regression and least absolute shrinkage and selection operator (LASSO) regression analyses were used to establish a risk prediction model and web-based risk calculator based on the training cohort data. The predictive accuracy of the model was verified in the validation cohort. Results We identified four variables as independent risk factors for brain WMD in neonates by multivariate logistic regression and LASSO analysis, including gestational age, fetal distress, prelabor rupture of membranes, and use of corticosteroids. These were used to establish a risk prediction nomogram and web-based calculator (https://caowenjun.shinyapps.io/dynnomapp/). The C-index of the training and validation sets was 0.898 (95% confidence interval: 0.8745-0.9215) and 0.887 (95% confidence interval: 0.8478-0.9262), respectively. Decision tree analysis showed that the model was highly effective in the threshold range of 1-61%. The sensitivity and specificity of the model were 82.5 and 81.7%, respectively, and the cutoff value was 0.099. Conclusion This is the first study describing the use of a nomogram and web-based calculator to predict the risk of WMD in neonates. The web-based calculator increases the applicability of the predictive model and is a convenient tool for doctors at primary hospitals and outpatient clinics, family doctors, and even parents to identify high-risk births early on and implementing appropriate interventions while avoiding excessive treatment of low-risk patients.
Collapse
Affiliation(s)
- Wenjun Cao
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Chenghan Luo
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengyuan Lei
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Shen
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenqian Ding
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mengmeng Wang
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Min Song
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jian Ge
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qian Zhang
- Neonatal Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
15
|
Du Z, Li G, Ge H, Zhou X, Zhang J. Design, Synthesis and Biological Evaluation of Steroidal Glycoconjugates as Potential Antiproliferative Agents. ChemMedChem 2021; 16:1488-1498. [PMID: 33476082 DOI: 10.1002/cmdc.202000966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/19/2021] [Indexed: 01/06/2023]
Abstract
To systematically evaluate the impact of neoglycosylation upon the anticancer activities and selectivity of steroids, four series of neoglycosides of diosgenin, pregnenolone, dehydroepiandrosterone and estrone were designed and synthesized according to the neoglycosylation approach. The structures of all the products were elucidated by NMR analysis, and the stereochemistry of C20-MeON-pregnenolone was confirmed by crystal X-ray diffraction. The compounds' cytotoxicity on five human cancer cell lines was evaluated using a Cell Counting Kit-8 assay, and structure-activity relationships (SAR) are discussed. 2-deoxy-d-glucoside 5 k displayed the most potent antiproliferative activities against HepG2 cells with an IC50 value of 1.5 μM. Further pharmacological experiments on compound 5 k on HepG2 cells revealed that it could cause morphological changes and cell-cycle arrest at the G0/G1 phase and then induced the apoptosis, which might be associated with the enhanced expression of high-mobility group Box 1 (HMGB1). Taken together, these findings prove that the neoglycosylation of steroids could be a promising strategy for the discovery of potential antiproliferative agents.
Collapse
Affiliation(s)
- Zhichao Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 210009, P. R. China
| | - Guolong Li
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, Jiangsu, 211198, P. R. China
| | - Haixia Ge
- School of Life Sciences, Huzhou University, Huzhou, Zhejiang, 313000, P. R. China
| | - Xiaoyang Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 210009, P. R. China
| | - Jian Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 210009, P. R. China.,ZhenPing Expert Workstation for Zhang Jian Zhenping, Ankang, Shaanxi, 725699, P. R. China
| |
Collapse
|
16
|
Löhndorf A, Hosang L, Dohle W, Odoardi F, Waschkowski SA, Rosche A, Bauche A, Winzer R, Tolosa E, Windhorst S, Marry S, Flügel A, Potter BVL, Diercks BP, Guse AH. 2-Methoxyestradiol and its derivatives inhibit store-operated Ca 2+ entry in T cells: Identification of a new and potent inhibitor. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118988. [PMID: 33581218 PMCID: PMC8062851 DOI: 10.1016/j.bbamcr.2021.118988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/29/2021] [Accepted: 02/04/2021] [Indexed: 12/15/2022]
Abstract
T cell activation starts with formation of second messengers that release Ca2+ from the endoplasmic reticulum (ER) and thereby activate store-operated Ca2+ entry (SOCE), one of the essential signals for T cell activation. Recently, the steroidal 2-methoxyestradiol was shown to inhibit nuclear translocation of the nuclear factor of activated T cells (NFAT). We therefore investigated 2-methoxyestradiol for inhibition of Ca2+ entry in T cells, screened a library of 2-methoxyestradiol analogues, and characterized the derivative 2-ethyl-3-sulfamoyloxy-17β-cyanomethylestra-1,3,5(10)-triene (STX564) as a novel, potent and specific SOCE inhibitor. STX564 inhibits Ca2+ entry via SOCE without affecting other ion channels and pumps involved in Ca2+ signaling in T cells. Downstream effects such as cytokine expression and cell proliferation were also inhibited by both 2-methoxyestradiol and STX564, which has potential as a new chemical biology tool.
Collapse
Affiliation(s)
- Anke Löhndorf
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Leon Hosang
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Wolfgang Dohle
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Francesca Odoardi
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Sissy-Alina Waschkowski
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Anette Rosche
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Andreas Bauche
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Riekje Winzer
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Sabine Windhorst
- Department of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Stephen Marry
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Alexander Flügel
- Institute for Neuroimmunology and Multiple Sclerosis Research, University Medical Centre Göttingen, Von-Siebold-Straße 3a, D-37075 Göttingen, Germany
| | - Barry V L Potter
- Drug Discovery & Medicinal Chemistry, Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, United Kingdom
| | - Björn-Philipp Diercks
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Andreas H Guse
- The Ca(2+) Signalling Group, Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany.
| |
Collapse
|
17
|
Stoll D, Yokota R, Sanches Aragão D, Casarini DE. Both aldosterone and spironolactone can modulate the intracellular ACE/ANG II/AT1 and ACE2/ANG (1-7)/MAS receptor axes in human mesangial cells. Physiol Rep 2020; 7:e14105. [PMID: 31165585 PMCID: PMC6548847 DOI: 10.14814/phy2.14105] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 04/10/2019] [Accepted: 04/24/2019] [Indexed: 12/01/2022] Open
Abstract
The kidney is an important target of the renin‐ANG‐aldosterone system (RAAS). To date, several studies have demonstrated the existence of a local RAAS in various tissues, including the renal tissue. The mineralocorticoid aldosterone is known to play a critical role in the classical RAAS; however, its effect on mesangial cells (MCs) remains to be elucidated. Based on this, our aim was to investigate whether aldosterone stimulation can modulate the intracellular RAAS of immortalized human MCs by evaluating ANG‐converting enzyme (ACE)/ANG II/ANG II receptor type 1 (AT1) and ANG‐converting enzyme 2 (ACE2)/ANG (1‐7)/MAS receptor axes. To realise this, protein expression, enzyme activity, and immunofluorescence were performed under aldosterone stimulation and in the presence of the mineralocorticoid receptor (MR) antagonist spironolactone (SPI). We observed that high doses of aldosterone increase ACE activity. The effect of aldosterone on the catalytic activity of ACE was completely abolished with the pretreatment of SPI suggesting that the aldosterone‐induced cell injuries through ANG II release were attenuated. Aldosterone treatment also decreased the expression of MAS receptor, but did not alter the expression or the catalytic activity of ACE 2 and ANG (1‐7) levels. Spironolactone modulated the localization of ANG II and AT1 receptor and decreased ANG (1‐7) and MAS receptor levels. Our data suggest that both aldosterone and the MR receptor antagonist can modulate both of these axes and that spironolactone can protect MCs from the damage induced by aldosterone.
Collapse
Affiliation(s)
- Danielle Stoll
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| | - Rodrigo Yokota
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| | - Danielle Sanches Aragão
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| | - Dulce E Casarini
- Escola Paulista de Medicina - Department of Medicine, Nephrology Division, Universidade Federal de São Paulo, UNIFESP, São Paulo, Brazil
| |
Collapse
|
18
|
Weiss SL, Peters MJ, Alhazzani W, Agus MSD, Flori HR, Inwald DP, Nadel S, Schlapbach LJ, Tasker RC, Argent AC, Brierley J, Carcillo J, Carrol ED, Carroll CL, Cheifetz IM, Choong K, Cies JJ, Cruz AT, De Luca D, Deep A, Faust SN, De Oliveira CF, Hall MW, Ishimine P, Javouhey E, Joosten KFM, Joshi P, Karam O, Kneyber MCJ, Lemson J, MacLaren G, Mehta NM, Møller MH, Newth CJL, Nguyen TC, Nishisaki A, Nunnally ME, Parker MM, Paul RM, Randolph AG, Ranjit S, Romer LH, Scott HF, Tume LN, Verger JT, Williams EA, Wolf J, Wong HR, Zimmerman JJ, Kissoon N, Tissieres P. Surviving sepsis campaign international guidelines for the management of septic shock and sepsis-associated organ dysfunction in children. Intensive Care Med 2020; 46:10-67. [PMID: 32030529 PMCID: PMC7095013 DOI: 10.1007/s00134-019-05878-6] [Citation(s) in RCA: 294] [Impact Index Per Article: 73.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To develop evidence-based recommendations for clinicians caring for children (including infants, school-aged children, and adolescents) with septic shock and other sepsis-associated organ dysfunction. DESIGN A panel of 49 international experts, representing 12 international organizations, as well as three methodologists and three public members was convened. Panel members assembled at key international meetings (for those panel members attending the conference), and a stand-alone meeting was held for all panel members in November 2018. A formal conflict-of-interest policy was developed at the onset of the process and enforced throughout. Teleconferences and electronic-based discussion among the chairs, co-chairs, methodologists, and group heads, as well as within subgroups, served as an integral part of the guideline development process. METHODS The panel consisted of six subgroups: recognition and management of infection, hemodynamics and resuscitation, ventilation, endocrine and metabolic therapies, adjunctive therapies, and research priorities. We conducted a systematic review for each Population, Intervention, Control, and Outcomes question to identify the best available evidence, statistically summarized the evidence, and then assessed the quality of evidence using the Grading of Recommendations Assessment, Development, and Evaluation approach. We used the evidence-to-decision framework to formulate recommendations as strong or weak, or as a best practice statement. In addition, "in our practice" statements were included when evidence was inconclusive to issue a recommendation, but the panel felt that some guidance based on practice patterns may be appropriate. RESULTS The panel provided 77 statements on the management and resuscitation of children with septic shock and other sepsis-associated organ dysfunction. Overall, six were strong recommendations, 49 were weak recommendations, and nine were best-practice statements. For 13 questions, no recommendations could be made; but, for 10 of these, "in our practice" statements were provided. In addition, 52 research priorities were identified. CONCLUSIONS A large cohort of international experts was able to achieve consensus regarding many recommendations for the best care of children with sepsis, acknowledging that most aspects of care had relatively low quality of evidence resulting in the frequent issuance of weak recommendations. Despite this challenge, these recommendations regarding the management of children with septic shock and other sepsis-associated organ dysfunction provide a foundation for consistent care to improve outcomes and inform future research.
Collapse
Affiliation(s)
- Scott L Weiss
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| | - Mark J Peters
- Great Ormond Street Hospital for Children, London, UK
| | - Waleed Alhazzani
- Department of Medicine, Division of Critical Care, McMaster University, Hamilton, ON, Canada
- Department of Health Research Methods and Impact, McMaster University, Hamilton, ON, Canada
| | - Michael S D Agus
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | - Luregn J Schlapbach
- Paediatric Critical Care Research Group, The University of Queensland and Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Robert C Tasker
- Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Andrew C Argent
- Red Cross War Memorial Children's Hospital and University of Cape Town, Cape Town, South Africa
| | - Joe Brierley
- Great Ormond Street Hospital for Children, London, UK
| | | | | | | | | | - Karen Choong
- Department of Medicine, Division of Critical Care, McMaster University, Hamilton, ON, Canada
- Department of Health Research Methods and Impact, McMaster University, Hamilton, ON, Canada
| | - Jeffry J Cies
- St. Christopher's Hospital for Children, Philadelphia, PA, USA
| | | | - Daniele De Luca
- Paris South University Hospitals-Assistance Publique Hopitaux de Paris, Paris, France
- Physiopathology and Therapeutic Innovation Unit-INSERM U999, South Paris-Saclay University, Paris, France
| | | | - Saul N Faust
- University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, UK
| | | | - Mark W Hall
- Nationwide Children's Hospital, Columbus, OH, USA
| | | | | | | | - Poonam Joshi
- All India Institute of Medical Sciences, New Delhi, India
| | - Oliver Karam
- Children's Hospital of Richmond at VCU, Richmond, VA, USA
| | | | - Joris Lemson
- Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Graeme MacLaren
- National University Health System, Singapore, Singapore
- Royal Children's Hospital, Melbourne, VIC, Australia
| | - Nilesh M Mehta
- Department of Anesthesiology, Critical Care and Pain, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | - Akira Nishisaki
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mark E Nunnally
- New York University Langone Medical Center, New York, NY, USA
| | | | - Raina M Paul
- Advocate Children's Hospital, Park Ridge, IL, USA
| | - Adrienne G Randolph
- Department of Anesthesiology, Critical Care and Pain, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Judy T Verger
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- College of Nursing, University of Iowa, Iowa City, IA, USA
| | | | - Joshua Wolf
- St. Jude Children's Research Hospital, Memphis, TN, USA
| | | | | | | | - Pierre Tissieres
- Paris South University Hospitals-Assistance Publique Hopitaux de Paris, Paris, France
- Institute of Integrative Biology of the Cell-CNRS, CEA, Univ Paris Sud, Gif-Sur-Yvette, France
| |
Collapse
|
19
|
Lorigo M, Mariana M, Lemos MC, Cairrao E. Vascular mechanisms of testosterone: The non-genomic point of view. J Steroid Biochem Mol Biol 2020; 196:105496. [PMID: 31655180 DOI: 10.1016/j.jsbmb.2019.105496] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/26/2019] [Accepted: 10/07/2019] [Indexed: 01/19/2023]
Abstract
Testosterone (T) is the predominant endogenous androgen in the bloodstream. At the vascular level, T presents genomic and non-genomic effects, and both effects may overlap. The genomic actions assume that androgens can freely cross the plasma membrane of target cells and bind to nuclear androgen receptors, inducing gene transcription and protein synthesis. The non-genomic effects have a more rapid onset and may be related to the interaction with protein/receptor/ion channels of the plasma membrane. The key T effect at the vascular level is vasorelaxation, which is primarily due to its rapid effect. Thus, the main purpose of this review is to discuss the T non-genomic effects at the vascular level and the molecular pathways involved in its vasodilator effect observed in in vivo and in vitro studies. In this sense, the nuclear receptor activation, the influence of vascular endothelium and the activation or inhibition of ion channels (potassium and calcium channels, respectively) will be reviewed regarding all the data that corroborated or not. Moreover, this review also provides a brief update on the association of T with the risk factors for cardiovascular diseases, namely metabolic syndrome, type 2 diabetes mellitus, obesity, atherosclerosis, dyslipidaemia, and hypertension. In summary, in this paper we consider the non-genomic vascular mode of action of androgen in physiological conditions and the main risk factors for cardiovascular diseases.
Collapse
Affiliation(s)
- Margarida Lorigo
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Melissa Mariana
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Manuel C Lemos
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Elisa Cairrao
- CICS-UBI - Centro de Investigação em Ciências da Saúde, University of Beira Interior, 6200-506 Covilhã, Portugal.
| |
Collapse
|
20
|
Weiss SL, Peters MJ, Alhazzani W, Agus MSD, Flori HR, Inwald DP, Nadel S, Schlapbach LJ, Tasker RC, Argent AC, Brierley J, Carcillo J, Carrol ED, Carroll CL, Cheifetz IM, Choong K, Cies JJ, Cruz AT, De Luca D, Deep A, Faust SN, De Oliveira CF, Hall MW, Ishimine P, Javouhey E, Joosten KFM, Joshi P, Karam O, Kneyber MCJ, Lemson J, MacLaren G, Mehta NM, Møller MH, Newth CJL, Nguyen TC, Nishisaki A, Nunnally ME, Parker MM, Paul RM, Randolph AG, Ranjit S, Romer LH, Scott HF, Tume LN, Verger JT, Williams EA, Wolf J, Wong HR, Zimmerman JJ, Kissoon N, Tissieres P. Surviving Sepsis Campaign International Guidelines for the Management of Septic Shock and Sepsis-Associated Organ Dysfunction in Children. Pediatr Crit Care Med 2020; 21:e52-e106. [PMID: 32032273 DOI: 10.1097/pcc.0000000000002198] [Citation(s) in RCA: 504] [Impact Index Per Article: 126.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To develop evidence-based recommendations for clinicians caring for children (including infants, school-aged children, and adolescents) with septic shock and other sepsis-associated organ dysfunction. DESIGN A panel of 49 international experts, representing 12 international organizations, as well as three methodologists and three public members was convened. Panel members assembled at key international meetings (for those panel members attending the conference), and a stand-alone meeting was held for all panel members in November 2018. A formal conflict-of-interest policy was developed at the onset of the process and enforced throughout. Teleconferences and electronic-based discussion among the chairs, co-chairs, methodologists, and group heads, as well as within subgroups, served as an integral part of the guideline development process. METHODS The panel consisted of six subgroups: recognition and management of infection, hemodynamics and resuscitation, ventilation, endocrine and metabolic therapies, adjunctive therapies, and research priorities. We conducted a systematic review for each Population, Intervention, Control, and Outcomes question to identify the best available evidence, statistically summarized the evidence, and then assessed the quality of evidence using the Grading of Recommendations Assessment, Development, and Evaluation approach. We used the evidence-to-decision framework to formulate recommendations as strong or weak, or as a best practice statement. In addition, "in our practice" statements were included when evidence was inconclusive to issue a recommendation, but the panel felt that some guidance based on practice patterns may be appropriate. RESULTS The panel provided 77 statements on the management and resuscitation of children with septic shock and other sepsis-associated organ dysfunction. Overall, six were strong recommendations, 52 were weak recommendations, and nine were best-practice statements. For 13 questions, no recommendations could be made; but, for 10 of these, "in our practice" statements were provided. In addition, 49 research priorities were identified. CONCLUSIONS A large cohort of international experts was able to achieve consensus regarding many recommendations for the best care of children with sepsis, acknowledging that most aspects of care had relatively low quality of evidence resulting in the frequent issuance of weak recommendations. Despite this challenge, these recommendations regarding the management of children with septic shock and other sepsis-associated organ dysfunction provide a foundation for consistent care to improve outcomes and inform future research.
Collapse
Affiliation(s)
- Scott L Weiss
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Mark J Peters
- Great Ormond Street Hospital for Children, London, United Kingdom
| | - Waleed Alhazzani
- Department of Medicine, Division of Critical Care, and Department of Health Research Methods and Impact, McMaster University, Hamilton, ON, Canada
| | - Michael S D Agus
- Department of Pediatrics (to Dr. Agus), Department of Anesthesiology, Critical Care and Pain (to Drs. Mehta and Randolph), Boston Children's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | - Luregn J Schlapbach
- Paediatric Critical Care Research Group, The University of Queensland and Queensland Children's Hospital, Brisbane, QLD, Australia
| | - Robert C Tasker
- Department of Pediatrics (to Dr. Agus), Department of Anesthesiology, Critical Care and Pain (to Drs. Mehta and Randolph), Boston Children's Hospital and Harvard Medical School, Boston, MA
| | - Andrew C Argent
- Red Cross War Memorial Children's Hospital and University of Cape Town, Cape Town, South Africa
| | - Joe Brierley
- Great Ormond Street Hospital for Children, London, United Kingdom
| | | | | | | | | | - Karen Choong
- Department of Medicine, Division of Critical Care, and Department of Health Research Methods and Impact, McMaster University, Hamilton, ON, Canada
| | - Jeffry J Cies
- St. Christopher's Hospital for Children, Philadelphia, PA
| | | | - Daniele De Luca
- Paris South University Hospitals-Assistance Publique Hopitaux de Paris, Paris, France.,Physiopathology and Therapeutic Innovation Unit-INSERM U999, South Paris-Saclay University, Paris, France
| | - Akash Deep
- King's College Hospital, London, United Kingdom
| | - Saul N Faust
- University Hospital Southampton NHS Foundation Trust and University of Southampton, Southampton, United Kingdom
| | | | - Mark W Hall
- Nationwide Children's Hospital, Columbus, OH
| | | | | | | | - Poonam Joshi
- All India Institute of Medical Sciences, New Delhi, India
| | - Oliver Karam
- Children's Hospital of Richmond at VCU, Richmond, VA
| | | | - Joris Lemson
- Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Graeme MacLaren
- National University Health System, Singapore, and Royal Children's Hospital, Melbourne, VIC, Australia
| | - Nilesh M Mehta
- Department of Pediatrics (to Dr. Agus), Department of Anesthesiology, Critical Care and Pain (to Drs. Mehta and Randolph), Boston Children's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | - Akira Nishisaki
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | | | | | | | - Adrienne G Randolph
- Department of Pediatrics (to Dr. Agus), Department of Anesthesiology, Critical Care and Pain (to Drs. Mehta and Randolph), Boston Children's Hospital and Harvard Medical School, Boston, MA
| | | | | | | | - Lyvonne N Tume
- University of the West of England, Bristol, United Kingdom
| | - Judy T Verger
- Children's Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.,College of Nursing, University of Iowa, Iowa City, IA
| | | | - Joshua Wolf
- St. Jude Children's Research Hospital, Memphis, TN
| | | | | | - Niranjan Kissoon
- British Columbia Children's Hospital, Vancouver, British Columbia, Canada
| | - Pierre Tissieres
- Paris South University Hospitals-Assistance Publique Hopitaux de Paris, Paris, France.,Institute of Integrative Biology of the Cell-CNRS, CEA, Univ Paris Sud, Gif-sur-Yvette, France
| |
Collapse
|
21
|
Dong YC, Chen ZZ, Clarke AR, Niu CY. Changes in Energy Metabolism Trigger Pupal Diapause Transition of Bactrocera minax After 20-Hydroxyecdysone Application. Front Physiol 2019; 10:1288. [PMID: 31736767 PMCID: PMC6831740 DOI: 10.3389/fphys.2019.01288] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 09/25/2019] [Indexed: 11/21/2022] Open
Abstract
Correct timing of diapause entry and exit is critical for a species' survival. While many aspects of insect diapause are well-studied, the mechanisms underlying diapause termination remain largely unknown. The Chinese citrus fly, Bactrocera minax, is a univoltine insect with an obligatory pupal diapause. The application of 20-hydroxyecdysone (20E) is known to terminate diapause in B. minax, and we used this approach, along with isobaric tags for relative and absolute quantitation technology, to determine the proteins associated with diapause termination in this fly. Among 2,258 identified proteins, 1,169 proteins significantly differed at 1, 2, and 5 days post-injection of 20E, compared with the solvent-injected control group. Functional annotation revealed that the majority of differentially expressed proteins were enriched in the core energy metabolism of amino acids, proteins, lipids, and carbohydrates as well as in signal transduction pathways including PPAR signaling, Calcium signaling, Glucagon signaling, VEGF signaling, Ras signaling, cGMP-PKG signaling, and cAMP signaling. A combined transcriptomic and proteomic analysis suggested the involvement of energy metabolism in the response of diapause transition. RNA interference experiments disclosed that a 20E injection triggers diapause termination probably through non-genomic actions, rather than nuclear receptor mediated genomic actions. Our results provide extensive proteomic resources for insect diapause transition and offer a potential for pest control by incapacitating the regulation of diapause termination either by breaking diapause prematurely or by delaying diapause termination to render diapausing individuals at a high risk of mortality.
Collapse
Affiliation(s)
- Yong-Cheng Dong
- Key Laboratory of Biology and Sustainable Management of Plant Diseases and Pests of Anhui Higher Education Institutes, College of Plant Protection, Anhui Agricultural University, Hefei, China.,Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhen-Zhong Chen
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Anthony R Clarke
- Faculty of Science and Technology, School of Earth, Environmental and Biological Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Chang-Ying Niu
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
22
|
A Selective Ligand for Estrogen Receptor Proteins Discriminates Rapid and Genomic Signaling. Cell Chem Biol 2019; 26:1692-1702.e5. [PMID: 31706983 DOI: 10.1016/j.chembiol.2019.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 09/05/2019] [Accepted: 10/18/2019] [Indexed: 12/27/2022]
Abstract
Estrogen exerts extensive and diverse effects throughout the body of women. In addition to the classical nuclear estrogen receptors (ERα and ERβ), the G protein-coupled estrogen receptor GPER is an important mediator of estrogen action. Existing ER-targeted therapeutic agents act as GPER agonists. Here, we report the identification of a small molecule, named AB-1, with the previously unidentified activity of high selectivity for binding classical ERs over GPER. AB-1 also possesses a unique functional activity profile as an agonist of transcriptional activity but an antagonist of rapid signaling through ERα. Our results define a class of small molecules that discriminate between the classical ERs and GPER, as well as between modes of signaling within the classical ERs. Such an activity profile, if developed into an ER antagonist, could represent an opportunity for the development of first-in-class nuclear hormone receptor-targeted therapeutics for breast cancer exhibiting reduced acquired and de novo resistance.
Collapse
|
23
|
|
24
|
Taguchi H, Oishi K, Shingu K, Matsumoto H, Masuzawa M. Intrathecal betamethasone for cancer pain: A study of its analgesic efficacy and safety. Acta Anaesthesiol Scand 2019; 63:659-667. [PMID: 30536525 PMCID: PMC6587555 DOI: 10.1111/aas.13305] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 10/30/2018] [Accepted: 10/31/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND A preliminary study has shown effective cancer pain relief by intrathecal betamethasone (ITB). However, further evidence is needed to support this new approach. METHODS Cancer patients with opioid-resistant pain received lumbar intrathecal administration of betamethasone 2 or 3 mg once a week for 28 days. Immediate and short-term analgesia (using a percentage pain reduction scale and a numerical rating scale, NRS) and long-term analgesia (using NRS) were assessed. Patients were classified into two groups according to the most painful site of metastasis: vertebral column and/or surrounding nerve plexus metastases (group A) and other metastases distal from the vertebral column (group B). RESULTS A total of 104 patients received ITB. Pain relief was observed not only in the lower half but also in the upper half of the body. The proportion of group A patients who experienced immediate analgesia was 81% (47/58), which was significantly greater than that of group B (P < 0.001). A decrease in NRS scores 1 day after ITB administration was observed in significantly more patients in group A than in group B (P < 0.001). Long-term analgesia was also recorded in a greater proportion of patients in group A than in group B in the 7-day (59%, 38/64 vs 6%, 2/33) and 28-day periods (71%, 40/56 vs 31%, 8/26) (P < 0.001). No adverse effects related to neurotoxicity were recorded. CONCLUSION Intrathecal injection of betamethasone produced analgesia for opioid-resistant cancer pain, and may be a potent therapeutic option for intolerable pain from vertebral column and/or surrounding nerve plexus metastases.
Collapse
Affiliation(s)
- Hitoshi Taguchi
- Department of Anesthesiology Kansai Medical University Medical Center Moriguchi Japan
| | - Keiko Oishi
- Department of Anesthesiology Kansai Medical University Medical Center Moriguchi Japan
| | - Koh Shingu
- Department of Anesthesiology Kansai Medical University Hirakata Japan
| | - Hideo Matsumoto
- Department of Anesthesiology Kansai Medical University Medical Center Moriguchi Japan
| | - Munehiro Masuzawa
- Department of Anesthesiology Kansai Medical University Medical Center Moriguchi Japan
| |
Collapse
|
25
|
Sabbadin C, Bordin L, Donà G, Manso J, Avruscio G, Armanini D. Licorice: From Pseudohyperaldosteronism to Therapeutic Uses. Front Endocrinol (Lausanne) 2019; 10:484. [PMID: 31379750 PMCID: PMC6657287 DOI: 10.3389/fendo.2019.00484] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 07/03/2019] [Indexed: 01/06/2023] Open
Abstract
Licorice has been used as a medicinal plant from 2.500 years. It shows a wide range of biological and pharmacological activities, including anti-inflammatory and immune regulatory actions. One of its most known effects is the induction of hypertension, and it can induce what appears to be pseudohyperaldosteronism, due to glycyrrhetinic acid, the main active component of the root. Glycyrrhetinic acid and metabolites block the 11 beta-hydroxysteroid dehydrogenase type 2 and also bind mineralocorticoid receptors directly, acting as agonists. However, other interesting therapeutic uses of licorice are linked to its anti-androgen and estrogen-like activity, especially in the treatment of polycystic ovary syndrome (PCOS) in conjunction with spironolactone therapy. In this brief review, we report the main features and possible therapeutic uses of this ancient plant.
Collapse
Affiliation(s)
- Chiara Sabbadin
- Endocrinology Unit, Department of Medicine, University Hospital of Padua, Padua, Italy
| | - Luciana Bordin
- Department of Molecular Medicine—Biological Chemistry, University of Padua, Padua, Italy
| | - Gabriella Donà
- Department of Molecular Medicine—Biological Chemistry, University of Padua, Padua, Italy
| | - Jacopo Manso
- Endocrinology Unit, Department of Medicine, University Hospital of Padua, Padua, Italy
| | - Giampiero Avruscio
- Angiology Unit, Department of Cardiac, Thoracic and Vascular Sciences, University Hospital of Padua, Padua, Italy
| | - Decio Armanini
- Endocrinology Unit, Department of Medicine, University Hospital of Padua, Padua, Italy
- *Correspondence: Decio Armanini
| |
Collapse
|
26
|
Heming N, Sivanandamoorthy S, Meng P, Bounab R, Annane D. Immune Effects of Corticosteroids in Sepsis. Front Immunol 2018; 9:1736. [PMID: 30105022 PMCID: PMC6077259 DOI: 10.3389/fimmu.2018.01736] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 07/13/2018] [Indexed: 12/29/2022] Open
Abstract
Sepsis, a life-threatening organ dysfunction, results from a dysregulated host response to invading pathogens that may be characterized by overwhelming systemic inflammation or some sort of immune paralysis. Sepsis remains a major cause of morbidity and mortality. Treatment is nonspecific and relies on source control and organ support. Septic shock, the most severe form of sepsis is associated with the highest rate of mortality. Two large multicentre trials, undertaken 15 years apart, found that the combination of hydrocortisone and fludrocortisone significantly reduces mortality in septic shock. The corticosteroids family is composed of several molecules that are usually characterized according to their glucocorticoid and mineralocorticoid power, relative to hydrocortisone. While the immune effects of glucocorticoids whether mediated or not by the intracellular glucocorticoid receptor have been investigated for several decades, it is only very recently that potential immune effects of mineralocorticoids via non-renal mineralocorticoid receptors have gained popularity. We reviewed the respective role of glucocorticoids and mineralocorticoids in counteracting sepsis-associated dysregulated immune systems.
Collapse
Affiliation(s)
- Nicholas Heming
- General Intensive Care Unit, Raymond Poincaré Hospital, Garches, France.,U1173 Laboratory Inflammation and Infection, University of Versailles SQY-Paris Saclay - INSERM, Montigny-Le-Bretonneux, France
| | | | - Paris Meng
- General Intensive Care Unit, Raymond Poincaré Hospital, Garches, France
| | - Rania Bounab
- General Intensive Care Unit, Raymond Poincaré Hospital, Garches, France
| | - Djillali Annane
- General Intensive Care Unit, Raymond Poincaré Hospital, Garches, France.,U1173 Laboratory Inflammation and Infection, University of Versailles SQY-Paris Saclay - INSERM, Montigny-Le-Bretonneux, France
| |
Collapse
|
27
|
Marczell I, Balogh P, Nyiro G, Kiss AL, Kovacs B, Bekesi G, Racz K, Patocs A. Membrane-bound estrogen receptor alpha initiated signaling is dynamin dependent in breast cancer cells. Eur J Med Res 2018; 23:31. [PMID: 29880033 PMCID: PMC5992704 DOI: 10.1186/s40001-018-0328-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 05/19/2018] [Indexed: 01/22/2023] Open
Abstract
Background Although membrane-associated estrogen receptors (mERs) have been known to play important role in steroid-induced signal transmission, we still know little about their function in the estrogen-induced proliferation of breast cancer cells. Methods In our current work we tried to separate membrane-initiated estrogen receptor signaling from the overall estrogenic effect in MCF-7 breast carcinoma cells. Re-analyzing expression data from multiple microarray experiments, we selected a set of key regulatory genes involved in proliferation regulation and estrogen signaling to monitor estrogen-induced transcription changes. We then compared these expression changes after 17β-estradiol and a membrane receptor selective estrogen–BSA treatment using quantitative real-time PCR. In order to follow receptor trafficking we used light and electron microscopy. Results Our quantitative real-time PCR results confirmed that the selective membrane receptor agonist, estrogen–BSA induces similarly pronounced expression changes regarding these genes as 17β-estradiol. Morphological study revealed that the membrane-bound form of classical estrogen receptor alpha is internalized after ligand binding via dynamin-dependent, caveola-mediated endocytosis. Inhibition of this internalization with dynamin inhibitor, dynasore practically abolished the regulatory effect of E2-BSA, suggesting that interaction and internalization with the scaffold protein is necessary for effective signaling. Conclusions The physiological role of plasma membrane estrogen receptor alpha is intensively studied, yet there are still several aspects of it to be resolved. The dynamin-dependent, ligand-mediated internalization of mERs seems to play an important role in estrogen signaling. Our results may serve as another example of how membrane initiated estrogen signaling and nuclear receptor initiated signaling overlap and form an intertwined system. Electronic supplementary material The online version of this article (10.1186/s40001-018-0328-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Istvan Marczell
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary
| | - Petra Balogh
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Gabor Nyiro
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary.,Molecular Medicine Research Group, Hungarian Academy of Sciences, Budapest, Szentkirályi str. 46., 1088, Hungary
| | - Anna L Kiss
- Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Balazs Kovacs
- Department of Aquaculture, Szent Istvan University, Godollo, Hungary
| | - Gabor Bekesi
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary
| | - Karoly Racz
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary.,Department of Human Morphology and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Attila Patocs
- 2nd Department of Medicine, Semmelweis University, Budapest, Szentkirályi utca 46., 1088, Hungary. .,HAS-SE 'Lendület' Hereditary Endocrine Tumors Research Group, Hungarian Academy of Sciences, Semmelweis University, Budapest, 46. Szentkiralyi str, 1088, Hungary. .,Department of Laboratory Medicine, Semmelweis University, Budapest, Nagyvárad sq 4, 1089, Hungary.
| |
Collapse
|
28
|
Liu A, Menon K. Contributions of a survey and retrospective cohort study to the planning of a randomised controlled trial of corticosteroids in the treatment of paediatric septic shock. Trials 2018; 19:283. [PMID: 29784051 PMCID: PMC5963179 DOI: 10.1186/s13063-018-2664-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/03/2018] [Indexed: 11/19/2022] Open
Abstract
Background Randomised controlled trials (RCTs) are challenging to conduct in a paediatric critical-care environment. Background work, including surveys and observational studies, is often used to determine disease estimates, sample sizes and design protocols when planning such RCTs. Our objective was to determine the necessity of performing a survey or a retrospective chart review or both when planning an RCT on corticosteroids in the treatment of paediatric septic shock. Methods We compared information on corticosteroid use for moderate to severe paediatric septic shock obtained from a survey of physician beliefs and stated practices with that obtained from a retrospective cohort study. The survey was conducted between February and March 2012 and the retrospective study included children from birth to 17 years of age admitted from January 2010 to June 2011. The survey and the retrospective study were conducted at four academic tertiary care centres in Canada. Results Survey responses from 23 physicians and retrospective data from 81 septic shock patients were included. The survey identified time to discontinuation of vasoactive infusions as the most feasible and clinically important outcome for an RCT on corticosteroids for paediatric septic shock. The retrospective chart review provided means and standard deviations for the suggested primary outcome, from which we could estimate sample sizes and justify the minimal clinically important difference. The survey found that physicians believe that patients with severe septic shock were most likely to benefit from corticosteroid administration but the majority stated they would be unwilling to randomise such patients, suggesting a lack of individual physician equipoise. The combined information from the survey and retrospective study suggested that enrolment of patients with moderate septic shock would be more feasible but that strategies would still have to be implemented to prevent open-label corticosteroid use. Conclusions The survey provided valuable information on the choice of primary outcome, target population and physician equipoise. The retrospective study provided estimates of patient numbers, the minimal clinically important difference, evidence for community equipoise and physician practice patterns. Strong consideration should be given to performing both types of studies prior to conducting RCTs in paediatric critical-care environments.
Collapse
Affiliation(s)
- Anna Liu
- University of Ottawa, Ottawa, K1H 8M5, Canada
| | - Kusum Menon
- University of Ottawa, Ottawa, K1H 8M5, Canada. .,Children's Hospital of Eastern Ontario, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada.
| |
Collapse
|
29
|
McEwen BS. Redefining neuroendocrinology: Epigenetics of brain-body communication over the life course. Front Neuroendocrinol 2018; 49:8-30. [PMID: 29132949 DOI: 10.1016/j.yfrne.2017.11.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 12/15/2022]
Abstract
The brain is the central organ of stress and adaptation to stress that perceives and determines what is threatening, as well as the behavioral and physiological responses to the stressor, and it does so somewhat differently in males and females. The expression of steroid hormone receptors throughout the brain has broadened the definition of 'neuroendocrinology' to include the reciprocal communication between the entire brain and body via hormonal and neural pathways. Mediated in part via systemic hormonal influences, the adult and developing brain possess remarkable structural and functional plasticity in response to stress, including neuronal replacement, dendritic remodeling, and synapse turnover. This article is both an account of an emerging field elucidating brain-body interactions at multiple levels, from molecules to social organization, as well as a personal account of my laboratory's role and, most importantly, the roles of trainees and colleagues, along with my involvement in interdisciplinary groups working on this topic.
Collapse
Affiliation(s)
- Bruce S McEwen
- Laboratory of Neuroendocrinology, The Rockefeller University, 1230 York Ave, New York, NY 10065, USA. http://www.rockefeller.edu/labheads/mcewen/mcewen-lab.php
| |
Collapse
|
30
|
Nadal A, Fuentes E, Ripoll C, Villar-Pazos S, Castellano-Muñoz M, Soriano S, Martinez-Pinna J, Quesada I, Alonso-Magdalena P. Extranuclear-initiated estrogenic actions of endocrine disrupting chemicals: Is there toxicology beyond paracelsus? J Steroid Biochem Mol Biol 2018; 176:16-22. [PMID: 28159674 DOI: 10.1016/j.jsbmb.2017.01.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 01/16/2017] [Accepted: 01/20/2017] [Indexed: 12/21/2022]
Abstract
Endocrine Disrupting Chemicals (EDCs), including bisphenol-A (BPA) do not act as traditional toxic chemicals inducing massive cell damage or death in an unspecific manner. EDCs can work upon binding to hormone receptors, acting as agonists, antagonists or modulators. Bisphenol-A displays estrogenic activity and, for many years it has been classified as a weak estrogen, based on the classic transcriptional action of estrogen receptors serving as transcription factors. However, during the last two decades our knowledge about estrogen signaling has advanced considerably. It is now accepted that estrogen receptors ERα and ERβ activate signaling pathways outside the nucleus which may or may not involve transcription. In addition, a new membrane estrogen receptor, GPER, has been proposed. Pharmacological and molecular evidence, along with results obtained in genetically modified mice, demonstrated that BPA, and its substitute BPS, are potent estrogens acting at nanomolar concentrations via extranuclear ERα, ERβ, and GPER. The different signaling pathways activated by BPA and BPS explain the well-known estrogenic effects of low doses of EDCs as well as non-monotonic dose-response relationships. These signaling pathways may help to explain the actions of EDCs with estrogenic activity in the etiology of different pathologies, including type-2 diabetes and obesity.
Collapse
Affiliation(s)
- Angel Nadal
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain.
| | - Esther Fuentes
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain; Departamento de Biología Aplicada, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Cristina Ripoll
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Sabrina Villar-Pazos
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Manuel Castellano-Muñoz
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Sergi Soriano
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Alicante, Spain
| | - Juan Martinez-Pinna
- Departamento de Fisiología, Genética y Microbiología, Universidad de Alicante, Alicante, Spain
| | - Ivan Quesada
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Instituto de Bioingeniería, Universidad Miguel Hernández de Elche, Alicante, Spain; Departamento de Biología Aplicada, Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Paloma Alonso-Magdalena
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, Universidad Miguel Hernández de Elche, Elche, Alicante, Spain; Departamento de Biología Aplicada, Universidad Miguel Hernández de Elche, Alicante, Spain
| |
Collapse
|
31
|
Zsarnovszky A, Kiss D, Jocsak G, Nemeth G, Toth I, Horvath TL. Thyroid hormone- and estrogen receptor interactions with natural ligands and endocrine disruptors in the cerebellum. Front Neuroendocrinol 2018; 48:23-36. [PMID: 28987779 DOI: 10.1016/j.yfrne.2017.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/06/2017] [Accepted: 10/04/2017] [Indexed: 10/18/2022]
Abstract
Although the effects of phytoestrogens on brain function is widely unknown, they are often regarded as "natural" and thus as harmless. However, the effects of phytoestrogens or environmental pollutants on brain function is underestimated. Estrogen (17beta-estradiol, E2) and thyroid hormones (THs) play pivotal roles in brain development. In the mature brain, these hormones regulate metabolism on cellular and organismal levels. Thus, E2 and THs do not only regulate the energy metabolism of the entire organism, but simultaneously also regulate important homeostatic parameters of neurons and glia in the CNS. It is, therefore, obvious that the mechanisms through which these hormones exert their effects are pleiotropic and include both intra- and intercellular actions. These hormonal mechanisms are versatile, and the experimental investigation of simultaneous hormone-induced mechanisms is technically challenging. In addition, the normal physiological settings of metabolic parameters depend on a plethora of interactions of the steroid hormones. In this review, we discuss conceptual and experimental aspects of the gonadal and thyroid hormones as they relate to in vitro models of the cerebellum.
Collapse
Affiliation(s)
- Attila Zsarnovszky
- Department of Animal Physiology and Animal Health, Faculty of Agricultural and Environmental Sciences, Szent István University, Páter Károly u. 1, H-2100 Gödöllő, Hungary; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - David Kiss
- Departments of Physiology and Biochemistry, University of Veterinary Medicine, Budapest 1078, Hungary
| | - Gergely Jocsak
- Departments of Physiology and Biochemistry, University of Veterinary Medicine, Budapest 1078, Hungary
| | - Gabor Nemeth
- Department of Obstetrics and Gynecology, University of Szeged, School of Medicine, Szeged, Hungary
| | - Istvan Toth
- Departments of Physiology and Biochemistry, University of Veterinary Medicine, Budapest 1078, Hungary
| | - Tamas L Horvath
- Department of Animal Physiology and Animal Health, Faculty of Agricultural and Environmental Sciences, Szent István University, Páter Károly u. 1, H-2100 Gödöllő, Hungary; Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Departments of Anatomy and Histology, University of Veterinary Medicine, Budapest 1078, Hungary.
| |
Collapse
|
32
|
Androgen receptor regulates SRC expression through microRNA-203. Oncotarget 2017; 7:25726-41. [PMID: 27028864 PMCID: PMC5041939 DOI: 10.18632/oncotarget.8366] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/07/2016] [Indexed: 12/20/2022] Open
Abstract
The SRC kinase has pivotal roles in multiple developmental processes and in tumor progression. An inverse relationship has been observed between androgen receptor (AR) activity and SRC signaling in advanced prostate cancer (PCa); however, the modulation of AR/SRC crosstalk that leads to metastatic PCa is unclear. Here, we showed that patients with high SRC levels displayed correspondingly low canonical AR gene signatures. Our results demonstrated that activated AR induced miR-203 and reduced SRC levels in PCa model systems. miR-203 directly binds to the 3′ UTR of SRC and regulates the stability of SRC mRNA upon AR activation. Moreover, we found that progressive PCa cell migration and growth were associated with a decrease in AR-regulated miR-203 and an increase in SRC. Relationships among AR, miR-203, and SRC were also confirmed in clinical datasets and specimens. We suggest that the induction of SRC results in increased PCa metastasis that is linked to the dysregulation of the AR signaling pathway through the inactivation of miR-203.
Collapse
|
33
|
Distinct hormonal regulation of two types of sexual dimorphism in submandibular gland of mice. Cell Tissue Res 2017; 371:261-272. [DOI: 10.1007/s00441-017-2719-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 10/15/2017] [Indexed: 12/16/2022]
|
34
|
Verma RP, Dasnadi S, Zhao Y, Chen HH. A comparative analysis of ante- and postnatal clinical characteristics of extremely premature neonates suffering from refractory and non-refractory hypotension: Is early clinical differentiation possible? Early Hum Dev 2017; 113:49-54. [PMID: 28750269 DOI: 10.1016/j.earlhumdev.2017.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/19/2017] [Accepted: 07/02/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND About 25% of hypotensive ELBW infants are refractory to intravascular volume expansion and inotropic drugs (VI) and require hydrocortisone (HC). Such neonates suffer from complications of prolonged hypotension and extended therapy with VI. ELBW infants with refractory hypotension (RH) are clinically and biochemically indistinguishable from those who respond to VI. OBJECTIVE Early identification and differentiation of ELBW infants susceptible to steroid dependent hypotension from those who respond to inotropic medications. METHODS In a retrospective study the ante- and postnatal clinical characteristics of ELBW infants who received hydrocortisone (HC) for refractory hypotension (RH) were compared to those who responded to volume-inotropes (VI). RESULTS Infants in HC group had lower birth weight (BW, 675±121g) and gestational age (GA, 25.1±1.3weeks) and higher mean airway pressure and oxygen requirements, all independent of antenatal steroid (ANS) exposure. The receipt of ANS (p 0.01) and occurrences of maternal diabetes mellitus (GDM, p 0.01) were lower in HC group. ANS (OR 0.5, 95% CI 0.2-0.9, p 0.01) and GDM (OR 0.3, 95% CI 0.09-0.9, p 0.04) reduced the risk for RH. HC group had higher risk for IVH (OR 2.1, 95% CI 1.02-4.2 p=0.04) which declined in the multivariate analysis. A trend towards lower risk of ventriculomegaly (VM) was noted in HC group (OR 0.3, 95% CI 0.1-1.1), which became significant after controlling for BW (OR 0.2 95% CI 0.07-0.9, p 0.04). Similar trend was noted for maternal hypertension. CONCLUSION Hypotension in ELBW infants who are ≤25wks of GA and unexposed to ANS and GDM is refractory to VI therapy. Such neonates may benefit from an initial therapy with, or earlier institution of hydrocortisone. The trend towards a higher risk for VM with VI therapy needs validation in future studies.
Collapse
Affiliation(s)
- Rita P Verma
- Nassau University Medical Center, Department of Pediatrics, Division of Neonatology, 2201 Hempstead Turnpike, East Meadow, NY 11554, United States.
| | - Shaeequa Dasnadi
- Houston Methodist Sugarland Nurseries, Department of Pediatrics Section of Neonatology, Baylor College of Medicine, 6621 Fannin Street # B 06104, Houston, TX 77030, United States.
| | - Yuan Zhao
- Department of Applied Mathematics and Statistics, Stony Brook University, Stony Brook, NY 11794, United States.
| | - Hegang H Chen
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, 113 Howard Hall, 660 W. Redwood Street, Baltimore, MD 21201, United States.
| |
Collapse
|
35
|
Liu C, Liao Y, Fan S, Fu X, Xiong J, Zhou S, Zou M, Wang J. G-Protein-Coupled Estrogen Receptor Antagonist G15 Decreases Estrogen-Induced Development of Non-Small Cell Lung Cancer. Oncol Res 2017; 27:283-292. [PMID: 28877783 PMCID: PMC7848463 DOI: 10.3727/096504017x15035795904677] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
G-protein-coupled estrogen receptor (GPER) was found to promote non-small cell lung cancer (NSCLC) by estrogen, indicating the potential necessity of inhibiting GPER by a selective antagonist. This study was performed to elucidate the function of GPER-selective inhibitor G15 in NSCLC development. Cytoplasmic GPER (cGPER) and nuclear GPER (nGPER) were detected by immunohistochemical analysis in NSCLC samples. The relation of GPER and estrogen receptor β (ERβ) expression and correlation between GPER, ERβ, and clinical factors were analyzed. The effects of activating GPER and function of G15 were analyzed in the proliferation of A549 and H1793 cell lines and development of urethane-induced adenocarcinoma. Overexpression of cGPER and nGPER was detected in 80.49% (120/150) and 52.00% (78/150) of the NSCLC samples. High expression of GPER was related with higher stages, poorer differentiation, and high expression of ERβ. The protein level of GPER in the A549 and H1793 cell lines was increased by treatment with E2, G1 (GPER agonist), or fulvestrant (Ful; ERβ antagonist) and decreased by G15. Administration with G15 reversed the E2- or G1-induced cell growth by inhibiting GPER. In urethane-induced adenocarcinoma mice, the number of tumor nodules and tumor index increased in the E2 or G1 group and decreased by treatment with G15. These findings demonstrate that using G15 to block GPER signaling may be considered as a new therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Changyu Liu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Yongde Liao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Sheng Fan
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Xiangning Fu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Jing Xiong
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Sheng Zhou
- Department of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Man Zou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| | - Jianmiao Wang
- Department of Respiratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, P.R. China
| |
Collapse
|
36
|
Abstract
Hypotension is a common problem in neonates with complex underlying pathophysiology. Although treatment of low blood pressure is common, clinicians must use all available information to target neonates with compromised perfusion. Pharmacotherapy should be tailored to the specific physiologic perturbations of the individual neonate. Dopamine is the most commonly utilized agent and may be the most appropriate agent for septic shock with low diastolic blood pressure. However, alternative therapies should be considered for other etiologies of hypotension, including milrinone and vasopressin for persistent pulmonary hypertension of the newborn and dobutamine for patent ductus arteriosus. Additional studies are required to refine the approach to neonatal hypotension and document the long-term outcomes of treated neonates.
Collapse
|
37
|
Ryu CS, Klein K, Zanger UM. Membrane Associated Progesterone Receptors: Promiscuous Proteins with Pleiotropic Functions - Focus on Interactions with Cytochromes P450. Front Pharmacol 2017; 8:159. [PMID: 28396637 PMCID: PMC5366339 DOI: 10.3389/fphar.2017.00159] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 03/13/2017] [Indexed: 12/22/2022] Open
Abstract
Membrane-associated progesterone receptors (MAPR) are a group of four rather small, partially homologous proteins, which share a similar non-covalent heme-binding domain that is related to cytochrome b5, a well-known functional interaction partner of microsomal cytochrome P450 (CYP) monooxygenase systems. Apart from their structural similarities the four proteins progesterone membrane component 1 (PGRMC1, also referred to as IZA, sigma-2 receptor, Dap1), PGRMC2, neudesin (NENF) and neuferricin (CYB5D2) display surprisingly divergent and multifunctional physiological properties related to cholesterol/steroid biosynthesis, drug metabolism and response, iron homeostasis, heme trafficking, energy metabolism, autophagy, apoptosis, cell cycle regulation, cell migration, neural functions, and tumorigenesis and cancer progression. The purpose of this mini-review is to briefly summarize the structural and functional properties of MAPRs with particular focus on their interactions with the CYP system. For PGRMC1, originally identified as a non-canonical progesterone-binding protein that mediates some immediate non-genomic actions of progesterone, available evidence indicates mainly activating interactions with steroidogenic CYPs including CYP11A1, CYP21A2, CYP17, CYP19, CYP51A1, and CYP61A1, while interactions with drug metabolizing CYPs including CYP2C2, CYP2C8, CYP2C9, CYP2E1, and CYP3A4 were either ineffective or slightly inhibitory. For the other MAPRs the evidence is so far less conclusive. We also point out that experimental limitations question some of the previous conclusions. Use of appropriate model systems should help to further clarify the true impact of these proteins on CYP-mediated metabolic pathways.
Collapse
Affiliation(s)
- Chang S Ryu
- Department of Molecular and Cell Biology, Dr. Margarete Fischer-Bosch-Institute of Clinical PharmacologyStuttgart, Germany; Eberhard-Karls-UniversityTübingen, Germany
| | - Kathrin Klein
- Department of Molecular and Cell Biology, Dr. Margarete Fischer-Bosch-Institute of Clinical PharmacologyStuttgart, Germany; Eberhard-Karls-UniversityTübingen, Germany
| | - Ulrich M Zanger
- Department of Molecular and Cell Biology, Dr. Margarete Fischer-Bosch-Institute of Clinical PharmacologyStuttgart, Germany; Eberhard-Karls-UniversityTübingen, Germany
| |
Collapse
|
38
|
LC–MS/MS simultaneous analysis of allopregnanolone, epiallopregnanolone, pregnanolone, dehydroepiandrosterone and dehydroepiandrosterone 3-sulfate in human plasma. Bioanalysis 2017; 9:527-539. [DOI: 10.4155/bio-2016-0262] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: Several neuropsychopharmacological properties have been attributed to the 3α-reduced pregnane steroids, allopregnanolone and pregnanolone, as well as to dehydroepiandrosterone sulfate because of their ability to modulate γ-aminobutyric acid (GABAA) receptors in the CNS. In order to understand better their role in several mechanisms in CNS, a new methodology is proposed to monitor these compounds in human plasma. Methodology & results: The analytes were first derivatized with 2-hydrazinopyridine and extracted from plasma using SPE. Then, the compounds were separated and detected by LC–MS/MS. A mobile phase of formic acid (0.1%) in water and methanol through a gradient of composition and a flow rate of 0.3 ml min-1 resulted in good separations of the analytes. Linear responses in wide range of concentrations and LOQs ranging from 10 (dehydroepiandrosterone 3-sulfate) to 40 pg ml-1 (dehydroepiandrosterone) were obtained in <9 min. The method proposed has been validated and then applied to monitor these neurosteroids in plasma samples from ten volunteers. Conclusion: For the first time, a straightforward and reliable method for the chromatographic separation of allopregnanolone, epiallopregnanolone and pregnanolone, as well as of dehydroepiandrosterone and dehydroepiandrosterone 3-sulfate was carried out, with optimal accuracy, sensitivity and specificity.
Collapse
|
39
|
Castoria G, Auricchio F, Migliaccio A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis. FASEB J 2016; 31:1289-1300. [PMID: 28031322 DOI: 10.1096/fj.201601047r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/19/2016] [Indexed: 01/11/2023]
Abstract
In this review, we focus on the role played by the protein partners of ligand-activated extranuclear androgen receptor (AR) in the final effects of hormone action, such as proliferation, migration, and neuritogenesis. The choice of AR partner, at least in part, depends on cell type. Androgen-activated receptor directly associates with cytoplasmic Src tyrosine kinase in epithelial cells, whereas in mesenchymal and neuronal cells, it prevalently interacts with filamin A. In the former, proliferation represents the final hormonal outcome, whereas in the latter, either migration or neuritogenesis, respectively, occurs. Furthermore, AR partner filamin A is replaced with Src when mesenchymal cells are stimulated with very low androgen concentrations. Consequently, the migratory effect is replaced by mitogenesis. Use of peptides that prevent receptor/partner assembly abolishes the effects that are dependent on their association and offers new therapeutic approaches to AR-related diseases. Perturbation of migration is often associated with metastatic spreading in cancer. In turn, cell cycle aberration causes tumors to grow faster, whereas toxic signaling triggers neurodegenerative events in the CNS. Here, we provide examples of new tools that interfere in rapid androgen effects, including migration, proliferation, and neuronal differentiation, together with their potential therapeutic applications in AR-dependent diseases-mainly prostate cancer and neurodegenerative disorders.-Castoria, G., Auricchio, F., Migliaccio, A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis.
Collapse
Affiliation(s)
- Gabriella Castoria
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Ferdinando Auricchio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Antimo Migliaccio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
40
|
Farhood Z, Lambert PR. The physiologic role of corticosteroids in Ménière's disease. Am J Otolaryngol 2016; 37:455-8. [PMID: 27221028 DOI: 10.1016/j.amjoto.2016.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/25/2016] [Indexed: 02/05/2023]
Abstract
Multiple options exist to manage Ménière's disease (MD), ranging from dietary modifications to ablative surgery. Corticosteroids (CS) have long been used to manage MD, but their exact mechanism for disease alleviation is relatively uncertain. Glucocorticoid receptors have been shown to exist in the human inner ear and several studies propose they influence mechanisms of blood flow, fluid regulation, and ion regulation, with recent evidence describing the latter two. Corticosteroids have been shown to upregulate aquaporins and ion channels in the inner ear, and may have a positive effect on labyrinthine blood flow. Additionally, processes have been described in genomic and non-genomic manners. This text will review the literature on the actions of CS on the inner ear relevant to MD.
Collapse
|
41
|
Sakkiah S, Ng HW, Tong W, Hong H. Structures of androgen receptor bound with ligands: advancing understanding of biological functions and drug discovery. Expert Opin Ther Targets 2016; 20:1267-82. [PMID: 27195510 DOI: 10.1080/14728222.2016.1192131] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Androgen receptor (AR) is a ligand-dependent transcription factor and a member of the nuclear receptor superfamily. It plays a vital role in male sexual development and regulates gene expression in various tissues, including prostate. Androgens are compounds that exert their biological effects via interaction with AR. Binding of androgens to AR initiates conformational changes in AR that affect binding of co-regulator proteins and DNA. AR agonists and antagonists are widely used in a variety of clinical applications (i.e. hypogonadism and prostate cancer therapy). AREAS COVERED This review provides a close look at structures of AR-ligand complexes and mutations in the receptor that have been revealed, discusses current challenges in the field, and sheds light on future directions. EXPERT OPINION AR is one of the primary targets for the treatment of prostate cancer, as AR antagonists inhibit prostate cancer growth. However, these drugs are not effective for long-term treatment and lead to castration-resistant prostate cancer. The structures of AR-ligand complexes are an invaluable scientific asset that enhances our understanding of biological functions and mechanisms of androgenic and anti-androgenic chemicals as well as promotes the discovery of superior drug candidates.
Collapse
Affiliation(s)
- Sugunadevi Sakkiah
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Hui Wen Ng
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Weida Tong
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| | - Huixiao Hong
- a Division of Bioinformatics and Biostatistics , National Center for Toxicological Research, U.S. Food and Drug Administration , Jefferson , AR , USA
| |
Collapse
|
42
|
Zarif JC, Miranti CK. The importance of non-nuclear AR signaling in prostate cancer progression and therapeutic resistance. Cell Signal 2016; 28:348-356. [PMID: 26829214 PMCID: PMC4788534 DOI: 10.1016/j.cellsig.2016.01.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Accepted: 01/28/2016] [Indexed: 01/22/2023]
Abstract
The androgen receptor (AR) remains the major oncogenic driver of prostate cancer, as evidenced by the efficacy of androgen deprivation therapy (ADT) in naïve patients, and the continued effectiveness of second generation ADTs in castration resistant disease. However, current ADTs are limited to interfering with AR ligand binding, either through suppression of androgen production or the use of competitive antagonists. Recent studies demonstrate 1) the expression of constitutively active AR splice variants that no longer depend on androgen, and 2) the ability of AR to signal in the cytoplasm independently of its transcriptional activity (non-genomic); thus highlighting the need to consider other ways to target AR. Herein, we review canonical AR signaling, but focus on AR non-genomic signaling, some of its downstream targets and how these effectors contribute to prostate cancer cell behavior. The goals of this review are to 1) re-highlight the continued importance of AR in prostate cancer as the primary driver, 2) discuss the limitations in continuing to use ligand binding as the sole targeting mechanism, 3) discuss the implications of AR non-genomic signaling in cancer progression and therapeutic resistance, and 4) address the need to consider non-genomic AR signaling mechanisms and pathways as a viable targeting strategy in combination with current therapies.
Collapse
Affiliation(s)
- Jelani C Zarif
- The James Buchanan Brady Urological Institute at The Johns Hopkins University School of Medicine Baltimore, MD 21287, United States
| | - Cindy K Miranti
- Lab of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, United States.
| |
Collapse
|
43
|
Abstract
Communication between cells in a multicellular organism occurs by the production of ligands (proteins, peptides, fatty acids, steroids, gases, and other low-molecular-weight compounds) that are either secreted by cells or presented on their surface, and act on receptors on, or in, other target cells. Such signals control cell growth, migration, survival, and differentiation. Signaling receptors can be single-span plasma membrane receptors associated with tyrosine or serine/threonine kinase activities, proteins with seven transmembrane domains, or intracellular receptors. Ligand-activated receptors convey signals into the cell by activating signaling pathways that ultimately affect cytosolic machineries or nuclear transcriptional programs or by directly translocating to the nucleus to regulate transcription.
Collapse
Affiliation(s)
- Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, SE-75124 Uppsala, Sweden
| | - Benson Lu
- The Salk Institute for Biological Studies, Gene Expression Laboratory, La Jolla, California 92037
| | - Ron Evans
- The Salk Institute for Biological Studies, Gene Expression Laboratory, La Jolla, California 92037
| | - J Silvio Gutkind
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892-4340
| |
Collapse
|
44
|
Zarif JC, Lamb LE, Schulz VV, Nollet EA, Miranti CK. Androgen receptor non-nuclear regulation of prostate cancer cell invasion mediated by Src and matriptase. Oncotarget 2016; 6:6862-76. [PMID: 25730905 PMCID: PMC4466655 DOI: 10.18632/oncotarget.3119] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 01/08/2015] [Indexed: 11/25/2022] Open
Abstract
Castration-resistant prostate cancers still depend on nuclear androgen receptor (AR) function despite their lack of dependence on exogenous androgen. Second generation anti-androgen therapies are more efficient at blocking nuclear AR; however resistant tumors still develop. Recent studies indicate Src is highly active in these resistant tumors. By manipulating AR activity in several different prostate cancer cell lines through RNAi, drug treatment, and the use of a nuclear-deficient AR mutant, we demonstrate that androgen acting on cytoplasmic AR rapidly stimulates Src tyrosine kinase via a non-genomic mechanism. Cytoplasmic AR, acting through Src enhances laminin integrin-dependent invasion. Active Matriptase, which cleaves laminin, is elevated within minutes after androgen stimulation, and is subsequently shed into the medium. Matriptase activation and shedding induced by cytoplasmic AR is dependent on Src. Concomitantly, CDCP1/gp140, a Matriptase and Src substrate that controls integrin-based migration, is activated. However, only inhibition of Matriptase, but not CDCP1, suppresses the AR/Src-dependent increase in invasion. Matriptase, present in conditioned medium from AR-stimulated cells, is sufficient to enhance invasion in the absence of androgen. Thus, invasion is stimulated by a rapid but sustained increase in Src activity, mediated non-genomically by cytoplasmic AR, leading to rapid activation and shedding of the laminin protease Matriptase.
Collapse
Affiliation(s)
- Jelani C Zarif
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA.,Cell and Molecular Biology Program, Michigan State University, East Lansing, MI 48824, USA
| | - Laura E Lamb
- Department of Urology, Beaumont Health System - Research Institute, Royal Oak, MI 48073, USA
| | - Veronique V Schulz
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Eric A Nollet
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA.,Van Andel Institute Graduate School, Grand Rapids, MI 49503, USA
| | - Cindy K Miranti
- Laboratory of Integrin Signaling and Tumorigenesis, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
45
|
Effects of various pharmacological agents on the function of norepinephrine transporter. J UOEH 2015; 37:33-42. [PMID: 25787100 DOI: 10.7888/juoeh.37.33] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The norepinephrine transporter is selectively expressed in noradrenergic nerve terminals, where it can exert spatial and temporal control over the action of norepinephrine. The norepinephrine transporter mediates the termination of neurotransmission via the reuptake of norepinephrine released into the extracellular milieu. In the present brief review, we report our recent studies about the effects of various pharmacological agents such as fasudil, nicotine, pentazocine, ketamine and genistein on norepinephrine transporter function.
Collapse
|
46
|
G protein-coupled estrogen receptor (GPER) mediates NSCLC progression induced by 17β-estradiol (E2) and selective agonist G1. Med Oncol 2015; 32:104. [PMID: 25744245 DOI: 10.1007/s12032-015-0558-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 02/28/2015] [Indexed: 12/28/2022]
Abstract
Estrogen classically drives lung cancer development via estrogen receptor β (ERβ). However, fulvestrant, an anti-estrogen-based endocrine therapeutic treatment, shows limited effects for non-small cell lung cancer (NSCLC) in phase II clinical trials. G protein-coupled estrogen receptor (GPER), a third estrogen receptor that binds to estrogen, has been found to be activated by fulvestrant, stimulating the progression of breast, endometrial, and ovarian cancers. We here demonstrated that cytoplasm-GPER (cGPER) (80.49 %) and nucleus-GPER (53.05 %) were detected by immunohistochemical analysis in NSCLC samples. cGPER expression was related to stages IIIA-IV, lymph node metastasis, and poorly differentiated NSCLC. Selective agonist G1 and 17β-estradiol (E2) promoted the GPER-mediated proliferation, invasion, and migration of NSCLC cells. Additionally, in vitro administration of E2 and G1 increased the number of tumor nodules, tumor grade, and tumor index in a urethane-induced adenocarcinoma model. Importantly, the pro-tumorigenic effects of GPER induced by E2 were significantly reduced by co-administering the GPER inhibitor G15 and the ERβ inhibitor fulvestrant, as compared to administering fulvestrant alone both in vitro and in vivo. Moreover, the phosphorylation of MAPK and Akt was involved in E2/G1-induced GPER activation. In conclusion, our results indicated that a pro-tumor function of GPER exists that mediated E2-/G1-dependent NSCLC progression and showed better efficiency regarding the co-targeting of GPER and ERβ, providing a rationale for further investigation of anti-estrogen clinical therapy.
Collapse
|
47
|
Ferrario CM, Schiffrin EL. Role of mineralocorticoid receptor antagonists in cardiovascular disease. Circ Res 2015; 116:206-13. [PMID: 25552697 PMCID: PMC4283558 DOI: 10.1161/circresaha.116.302706] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 10/08/2014] [Indexed: 12/23/2022]
Abstract
Aldosterone exerts its best known sodium homeostasis actions by controlling sodium excretion at the level of the distal tubules via activation of the apical epithelial sodium channel and the basolateral Na(+)/K(+)ATPase pump. Recently, this mineralocorticoid hormone has been demonstrated to act on the heart and blood vessels. Excess release of aldosterone in relation to the salt status induces both genomic and nongenomic effects that by promoting endothelial dysfunction, and vascular and cardiorenal adverse remodeling, contribute to the target organ damage found in hypertension, heart failure, myocardial infarction, and chronic renal failure. Mineralocorticoid receptor blockers have been shown to be highly effective in resistant hypertension and to slow down heart failure progression, and in experimental animals, the development of atherosclerosis. Blockade of the action of aldosterone and potentially other mineralocorticoid steroids has been increasingly demonstrated to be an extremely beneficial therapy in different forms of cardiovascular disease. This review provides a summary of the knowledge that exists on aldosterone actions in the cardiovascular system and, in providing the translational impact of this knowledge to the clinical arena, illustrates how much more needs to be achieved in exploring the use of mineralocorticoid receptor blockers in less advanced stages of heart, renal, and vascular disease.
Collapse
Affiliation(s)
- Carlos M Ferrario
- From the Hypertension Translational Research Laboratory, Departments of Surgery, Internal Medicine-Nephrology, and Physiology-Pharmacology, Wake Forest University Health Science Center, Winston Salem, NC (C.M.F.); and Department of Medicine and Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, McGill University, Montreal, PQ, Canada (E.L.S.).
| | - Ernesto L Schiffrin
- From the Hypertension Translational Research Laboratory, Departments of Surgery, Internal Medicine-Nephrology, and Physiology-Pharmacology, Wake Forest University Health Science Center, Winston Salem, NC (C.M.F.); and Department of Medicine and Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, McGill University, Montreal, PQ, Canada (E.L.S.)
| |
Collapse
|
48
|
Ménard J, Rigel DF, Watson C, Jeng AY, Fu F, Beil M, Liu J, Chen W, Hu CW, Leung-Chu J, LaSala D, Liang G, Rebello S, Zhang Y, Dole WP. Aldosterone synthase inhibition: cardiorenal protection in animal disease models and translation of hormonal effects to human subjects. J Transl Med 2014; 12:340. [PMID: 25491597 PMCID: PMC4301837 DOI: 10.1186/s12967-014-0340-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/22/2014] [Indexed: 01/31/2023] Open
Abstract
Background Aldosterone synthase inhibition provides the potential to attenuate both the mineralocorticoid receptor-dependent and independent actions of aldosterone. In vitro studies with recombinant human enzymes showed LCI699 to be a potent, reversible, competitive inhibitor of aldosterone synthase (Ki = 1.4 ± 0.2 nmol/L in humans) with relative selectivity over 11β-hydroxylase. Methods Hormonal effects of orally administered LCI699 were examined in rat and monkey in vivo models of adrenocorticotropic hormone (ACTH) and angiotensin-II-stimulated aldosterone release, and were compared with the mineralocorticoid receptor antagonist eplerenone in a randomized, placebo-controlled study conducted in 99 healthy human subjects. The effects of LCI699 and eplerenone on cardiac and renal sequelae of aldosterone excess were investigated in a double-transgenic rat (dTG rat) model overexpressing human renin and angiotensinogen. Results Rat and monkey in vivo models of stimulated aldosterone release predicted human dose– and exposure–response relationships, but overestimated the selectivity of LCI699 in humans. In the dTG rat model, LCI699 dose-dependently blocked increases in aldosterone, prevented development of cardiac and renal functional abnormalities independent of blood pressure changes, and prolonged survival. Eplerenone prolonged survival to a similar extent, but was less effective in preventing cardiac and renal damage. In healthy human subjects, LCI699 0.5 mg selectively reduced plasma and 24 h urinary aldosterone by 49 ± 3% and 39 ± 6% respectively (Day 1, mean ± SEM; P < 0.001 vs placebo), which was associated with natriuresis and an increase in plasma renin activity. Doses of LCI699 greater than 1 mg inhibited basal and ACTH-stimulated cortisol. Eplerenone 100 mg increased plasma and 24 h urinary aldosterone while stimulating natriuresis and increasing renin activity. In contrast to eplerenone, LCI699 increased the aldosterone precursor 11-deoxycorticosterone and urinary potassium excretion. Conclusions These results provide new insights into the cardiac and renal effects of inhibiting aldosterone synthase in experimental models and translation of the hormonal effects to humans. Selective inhibition of aldosterone synthase appears to be a promising approach to treat diseases associated with aldosterone excess. Electronic supplementary material The online version of this article (doi:10.1186/s12967-014-0340-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joël Ménard
- Université Paris Descartes, Faculté de Médecine and INSERM/AP-HP Clinical Investigation Center, Georges Pompidou Hospital, Paris, France.
| | - Dean F Rigel
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Catherine Watson
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Arco Y Jeng
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA. .,Current address: Golda Och Academy, 1418 Pleasant Valley Way, West Orange, NJ, 07052, USA.
| | - Fumin Fu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Michael Beil
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Jing Liu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Wei Chen
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Chii-Whei Hu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | | | - Daniel LaSala
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Guiqing Liang
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| | - Sam Rebello
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - Yiming Zhang
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA.
| | - William P Dole
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA.
| |
Collapse
|
49
|
Asuthkar S, Demirkhanyan L, Sun X, Elustondo PA, Krishnan V, Baskaran P, Velpula KK, Thyagarajan B, Pavlov EV, Zakharian E. The TRPM8 protein is a testosterone receptor: II. Functional evidence for an ionotropic effect of testosterone on TRPM8. J Biol Chem 2014; 290:2670-88. [PMID: 25480785 DOI: 10.1074/jbc.m114.610873] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Testosterone is a key steroid hormone in the development of male reproductive tissues and the regulation of the central nervous system. The rapid signaling mechanism induced by testosterone affects numerous behavioral traits, including sexual drive, aggressiveness, and fear conditioning. However, the currently identified testosterone receptor(s) is not believed to underlie the fast signaling, suggesting an orphan pathway. Here we report that an ion channel from the transient receptor potential family, TRPM8, commonly known as the cold and menthol receptor is the major component of testosterone-induced rapid actions. Using cultured and primary cell lines along with the purified TRPM8 protein, we demonstrate that testosterone directly activates TRPM8 channel at low picomolar range. Specifically, testosterone induced TRPM8 responses in primary human prostate cells, PC3 prostate cancer cells, dorsal root ganglion neurons, and hippocampal neurons. Picomolar concentrations of testosterone resulted in full openings of the purified TRPM8 channel in planar lipid bilayers. Furthermore, acute applications of testosterone on human skin elicited a cooling sensation. Our data conclusively demonstrate that testosterone is an endogenous and highly potent agonist of TRPM8, suggesting a role of TRPM8 channels well beyond their well established function in somatosensory neurons. This discovery may further imply TRPM8 channel function in testosterone-dependent behavioral traits.
Collapse
Affiliation(s)
- Swapna Asuthkar
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Lusine Demirkhanyan
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Xiaohui Sun
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | | | - Vivek Krishnan
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Padmamalini Baskaran
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Kiran Kumar Velpula
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605
| | - Baskaran Thyagarajan
- the College of Health Sciences, School of Pharmacy, University of Wyoming, Laramie, Wyoming 82071, and
| | - Evgeny V Pavlov
- Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada, the Department of Basic Sciences, College of Dentistry, New York University, New York, New York 10010
| | - Eleonora Zakharian
- From the Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine, Peoria, Illinois 61605,
| |
Collapse
|
50
|
Boardman C, Chachi L, Gavrila A, Keenan CR, Perry MM, Xia YC, Meurs H, Sharma P. Mechanisms of glucocorticoid action and insensitivity in airways disease. Pulm Pharmacol Ther 2014; 29:129-43. [PMID: 25218650 DOI: 10.1016/j.pupt.2014.08.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 08/18/2014] [Accepted: 08/25/2014] [Indexed: 01/04/2023]
Abstract
Glucocorticoids are the mainstay for the treatment of chronic inflammatory diseases including asthma and chronic obstructive pulmonary disease (COPD). However, it has been recognized that glucocorticoids do not work well in certain patient populations suggesting reduced sensitivity. The ultimate biologic responses to glucocorticoids are determined by not only the concentration of glucocorticoids but also the differences between individuals in glucocorticoid sensitivity, which is influenced by multiple factors. Studies are emerging to understand these mechanisms in detail, which would help in increasing glucocorticoid sensitivity in patients with chronic airways disease. This review aims to highlight both classical and emerging concepts of the anti-inflammatory mechanisms of glucocorticoids and also review some novel strategies to overcome steroid insensitivity in airways disease.
Collapse
Affiliation(s)
- C Boardman
- Airway Disease, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - L Chachi
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - A Gavrila
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - C R Keenan
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - M M Perry
- Airway Disease, National Heart and Lung Institute, Imperial College, London, United Kingdom
| | - Y C Xia
- Department of Pharmacology, University of Melbourne, Parkville, Victoria, Australia
| | - H Meurs
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands
| | - P Sharma
- Department of Physiology and Pharmacology, Airways Inflammation Research Group, Snyder Institute for Chronic Diseases, University of Calgary, 4C46 HRIC, 3280 Hospital Dr NW, Calgary, AB, Canada T2N 4N1.
| |
Collapse
|