1
|
Morris MT, Pascoe JL, Busada JT. In vitro to in vivo evidence for chemical disruption of glucocorticoid receptor signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.07.642104. [PMID: 40161699 PMCID: PMC11952347 DOI: 10.1101/2025.03.07.642104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Glucocorticoids are steroid hormones that regulate stress homeostasis, metabolism, and inflammatory responses. Dysregulation of the glucocorticoid receptor (GR) is linked to diseases such as obesity, mood disorders, and immune dysfunction. Endocrine-disrupting chemicals (EDCs) are widespread environmental contaminants known to interfere with hormone signaling, but their impact on glucocorticoid signaling remains unclear. While several GR-disrupting compounds have been identified in vitro , their in vivo effects remain largely unknown. In this study, we identified the agricultural agents dichlorodiphenyltrichloroethane (DDT) and ziram as GR-disruptors in vitro. In vivo , corticosterone co-treatment with DDT or the GR antagonist RU-486 inhibited the expression of classic GR-regulated transcripts in the liver. Furthermore, chronic exposure to DDT or RU-486 significantly reduced circulating B and T lymphocyte populations, respectively. These findings underscore the need to translate in vitro discoveries into in vivo models to assess the clinical relevance of GR-disrupting compounds. Moreover, they highlight the potential for xenobiotic-induced GR disruption to impair metabolic and immune homeostasis, potentially increasing disease susceptibility.
Collapse
|
2
|
Im JH, Oh G, Fu X, Lim JS, Choi SI, Lee OH. Research status of anti-obesogenic functional foods: mechanism of endocrine-disrupting chemicals and glucocorticoid receptor pathway. Food Sci Biotechnol 2025; 34:829-835. [PMID: 39974849 PMCID: PMC11832854 DOI: 10.1007/s10068-024-01723-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/12/2024] [Accepted: 09/25/2024] [Indexed: 02/21/2025] Open
Abstract
Obesity due to excessive fat accumulation, affects health and quality of life and increases the risk of diseases such as type 2 diabetes and cardiovascular conditions. Traditional causes, such as calorie excess and sedentary behavior, do not fully explain the obesity epidemic, leading to the hypothesis that endocrine-disrupting chemicals or obesogens contribute to obesity. The obesogenic mechanisms of representative obesogenic substances, such as bisphenols, have been discussed, mainly focusing on their interactions with estrogen receptors. Based on several studies showing that obesogens induce obesity by mimicking glucocorticoids, this review focused on the role of the glucocorticoid receptor pathway. In addition, the anti-obesogenic bioactive substances that have been studied to this date along with their inhibitory mechanisms were discussed.
Collapse
Affiliation(s)
- Ji-Hyun Im
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon, 24341 South Korea
| | - Geon Oh
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon, 24341 South Korea
| | - Xiaolu Fu
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon, 24341 South Korea
| | - June Seok Lim
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon, 24341 South Korea
| | - Sun-Il Choi
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon, 24341 South Korea
| | - Ok-Hwan Lee
- Department of Food Biotechnology and Environmental Science, Kangwon National University, Chuncheon, 24341 South Korea
| |
Collapse
|
3
|
Clark AB, Conzen SD. Glucocorticoid receptor-mediated oncogenic activity is dependent on breast cancer subtype. J Steroid Biochem Mol Biol 2024; 243:106518. [PMID: 38734115 DOI: 10.1016/j.jsbmb.2024.106518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 03/23/2024] [Accepted: 04/08/2024] [Indexed: 05/13/2024]
Abstract
Breast cancer incidence has been steadily rising and is the leading cause of cancer death in women due to its high metastatic potential. Individual breast cancer subtypes are classified by both cell type of origin and receptor expression, namely estrogen, progesterone and human epidermal growth factor receptors (ER, PR and HER2). Recently, the importance and context-dependent role of glucocorticoid receptor (GR) expression in the natural history and prognosis of breast cancer subtypes have been uncovered. In ER-positive breast cancer, GR expression is associated with a better prognosis as a result of ER-GR crosstalk. GR appears to modulate ER-mediated gene expression resulting in decreased tumor cell proliferation and a more indolent cancer phenotype. In ER-negative breast cancer, including GR-positive triple-negative breast cancer (TNBC), GR expression enhances migration, chemotherapy resistance and cell survival. In invasive lobular carcinoma, GR function is relatively understudied, and more work is required to determine whether lobular subtypes behave similarly to their invasive ductal carcinoma counterparts. Importantly, understanding GR signaling in individual breast cancer subtypes has potential clinical implications because of the recent development of highly selective GR non-steroidal ligands, which represent a therapeutic approach for modulating GR activity systemically.
Collapse
Affiliation(s)
- Abigail B Clark
- Depatment of Internal Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Suzanne D Conzen
- Depatment of Internal Medicine, Division of Hematology and Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
4
|
Erradhouani C, Bortoli S, Aït‐Aïssa S, Coumoul X, Brion F. Metabolic disrupting chemicals in the intestine: the need for biologically relevant models: Zebrafish: what can we learn from this small environment-sensitive fish? FEBS Open Bio 2024; 14:1397-1419. [PMID: 39218795 PMCID: PMC11492336 DOI: 10.1002/2211-5463.13878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 07/08/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Although the concept of endocrine disruptors first appeared almost 30 years ago, the relatively recent involvement of these substances in the etiology of metabolic pathologies (obesity, diabetes, hepatic steatosis, etc.) has given rise to the concept of Metabolic Disrupting Chemicals (MDCs). Organs such as the liver and adipose tissue have been well studied in the context of metabolic disruption by these substances. The intestine, however, has been relatively unexplored despite its close link with these organs. In vivo models are useful for the study of the effects of MDCs in the intestine and, in addition, allow investigations into interactions with the rest of the organism. In the latter respect, the zebrafish is an animal model which is used increasingly for the characterization of endocrine disruptors and its use as a model for assessing effects on the intestine will, no doubt, expand. This review aims to highlight the importance of the intestine in metabolism and present the zebrafish as a relevant alternative model for investigating the effect of pollutants in the intestine by focusing, in particular, on cytochrome P450 3A (CYP3A), one of the major molecular players in endogenous and MDCs metabolism in the gut.
Collapse
Affiliation(s)
- Chedi Erradhouani
- Ecotoxicologie des Substances et des MilieuxINERISVerneuil‐en‐HalatteFrance
- Université Paris CitéFrance
- Inserm UMR‐S 1124ParisFrance
| | | | - Selim Aït‐Aïssa
- Ecotoxicologie des Substances et des MilieuxINERISVerneuil‐en‐HalatteFrance
| | | | - François Brion
- Ecotoxicologie des Substances et des MilieuxINERISVerneuil‐en‐HalatteFrance
| |
Collapse
|
5
|
Rahman A, Haider MF. A comprehensive review on glucocorticoids induced osteoporosis: A medication caused disease. Steroids 2024; 207:109440. [PMID: 38754651 DOI: 10.1016/j.steroids.2024.109440] [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: 03/27/2024] [Revised: 05/11/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Glucocorticoids (GCs) are steroid hormones that are extensively used in the treatment of autoimmune diseases, inflammation, and cancer. The major ill effect of administering GCs is that it has a deleterious effect on bone, which leads to GC-induced osteoporosis. GC therapy induces bone loss and is associated with the risk of nonvertebral and vertebral fractures, as it works in combination by increasing bone reabsorption and suppressing bone formation during the initial phase of therapy. It is seen and established that GC in excess or in low dose for 3 months or more can be a risk factor for fracture, and the risk increases with an increase in dose and duration of usage. The most common cause of secondary osteoporosis is the administration of GC inside the body to treat various diseases. The degree of bone loss is directly proportional to the GC dose and the exposure duration. The first step is to evaluate the patients' risk factors for the development of glucocorticoids that induce osteoporosis, which include the dose, duration of use, patient age, sex, previous fractures, and other medical conditions.
Collapse
Affiliation(s)
- Asim Rahman
- Faculty of Pharmacy, Integral University, Lucknow 226026, India
| | - Md Faheem Haider
- Faculty of Pharmacy, Integral University, Lucknow 226026, India.
| |
Collapse
|
6
|
Zhang Q, Yang Y, Liu J, Wu Y, Liu Y, Zhang J. Testicular dysfunction and "its recovery effect" after cadmium exposure. Food Chem Toxicol 2024; 188:114656. [PMID: 38615797 DOI: 10.1016/j.fct.2024.114656] [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: 03/07/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024]
Abstract
In recent years, with the acceleration of industrialization, the decline of male fertility caused by heavy metal pollution has attracted much attention. However, whether the inhibition of testicular function after cadmium exposure is reversible remains to be studied. In this study, we constructed rat models of cadmium exposure and dis-exposure, and collected relative samples to observe the changes of related indicators. The results showed that cadmium exposure could reduce the fertility, inhibit the hypothalamic-pituitary-testis axis and activate hypothalamic-pituitary-adrenal axis function, the testicular GR/PI3K-AKT/AMPK signal was abnormal, cell proliferation was inhibited and apoptosis was enhanced. Four weeks after the exposure was stopped, the fertility was still decreased, testicular testosterone synthesis and spermatogenesis were inhibited, cell proliferation was inhibited and apoptosis was enhanced, but all of them were reversed. After eight weeks of cadmium exposure, the above indicators were observed to return to normal. At the same time, by giving different concentrations of corticosterone to spermatogonium, we confirmed that corticosterone may regulate the proliferation and apoptosis of spermatogonium through GR/PI3K-AKT/AMPK signal. In this study, the reproductive toxicity of cadmium, a metal environmental pollutant, was analyzed in depth to provide a new theoretical and experimental basis for ensuring male reproductive health.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China
| | - YanLing Yang
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China
| | - Juan Liu
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China
| | - YuJiao Wu
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China
| | - Yi Liu
- WuHan University, WuHan, Hubei, 430070, China.
| | - Jing Zhang
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China.
| |
Collapse
|
7
|
de Zoysa N, Haruhara K, Nikolic-Paterson DJ, Kerr PG, Ling J, Gazzard SE, Puelles VG, Bertram JF, Cullen-McEwen LA. Podocyte number and glomerulosclerosis indices are associated with the response to therapy for primary focal segmental glomerulosclerosis. Front Med (Lausanne) 2024; 11:1343161. [PMID: 38510448 PMCID: PMC10951056 DOI: 10.3389/fmed.2024.1343161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/19/2024] [Indexed: 03/22/2024] Open
Abstract
Corticosteroid therapy, often in combination with inhibition of the renin-angiotensin system, is first-line therapy for primary focal and segmental glomerulosclerosis (FSGS) with nephrotic-range proteinuria. However, the response to treatment is variable, and therefore new approaches to indicate the response to therapy are required. Podocyte depletion is a hallmark of early FSGS, and here we investigated whether podocyte number, density and/or size in diagnostic biopsies and/or the degree of glomerulosclerosis could indicate the clinical response to first-line therapy. In this retrospective single center cohort study, 19 participants (13 responders, 6 non-responders) were included. Biopsies obtained at diagnosis were prepared for analysis of podocyte number, density and size using design-based stereology. Renal function and proteinuria were assessed 6 months after therapy commenced. Responders and non-responders had similar levels of proteinuria at the time of biopsy and similar kidney function. Patients who did not respond to treatment at 6 months had a significantly higher percentage of glomeruli with global sclerosis than responders (p < 0.05) and glomerulosclerotic index (p < 0.05). Podocyte number per glomerulus in responders was 279 (203-507; median, IQR), 50% greater than that of non-responders (186, 118-310; p < 0.05). These findings suggest that primary FSGS patients with higher podocyte number per glomerulus and less advanced glomerulosclerosis are more likely to respond to first-line therapy at 6 months. A podocyte number less than approximately 216 per glomerulus, a GSI greater than 1 and percentage global sclerosis greater than approximately 20% are associated with a lack of response to therapy. Larger, prospective studies are warranted to confirm whether these parameters may help inform therapeutic decision making at the time of diagnosis of primary FSGS.
Collapse
Affiliation(s)
- Natasha de Zoysa
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Kotaro Haruhara
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- Division of Nephrology and Hypertension, Jikei University School of Medicine, Tokyo, Japan
| | - David J. Nikolic-Paterson
- Department of Nephrology, Monash Medical Centre, Clayton, VIC, Australia
- Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Peter G. Kerr
- Department of Nephrology, Monash Medical Centre, Clayton, VIC, Australia
- Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Jonathan Ling
- Department of Nephrology, Monash Medical Centre, Clayton, VIC, Australia
- Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Sarah E. Gazzard
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| | - Victor G. Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | - John F. Bertram
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Melbourne, VIC, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Brisbane, QLD, Australia
| | - Luise A. Cullen-McEwen
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Clayton, VIC, Australia
| |
Collapse
|
8
|
Mata-Greenwood E, Chow WL, Anti NAO, Sands LD, Adeoye O, Ford SP, Nathanielsz PW. Dysregulation of Glucocorticoid Receptor Homeostasis and Glucocorticoid-Associated Genes in Umbilical Cord Endothelial Cells of Diet-Induced Obese Pregnant Sheep. Int J Mol Sci 2024; 25:2311. [PMID: 38396987 PMCID: PMC10888705 DOI: 10.3390/ijms25042311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Maternal obesity (MO) is associated with offspring cardiometabolic diseases that are hypothesized to be partly mediated by glucocorticoids. Therefore, we aimed to study fetal endothelial glucocorticoid sensitivity in an ovine model of MO. Rambouillet/Columbia ewes were fed either 100% (control) or 150% (MO) National Research Council recommendations from 60 d before mating until near-term (135 days gestation). Sheep umbilical vein and artery endothelial cells (ShUVECs and ShUAECs) were used to study glucocorticoid receptor (GR) expression and function in vitro. Dexamethasone dose-response studies of gene expression, activation of a glucocorticoid response element (GRE)-dependent luciferase reporter vector, and cytosolic/nuclear GR translocation were used to assess GR homeostasis. MO significantly increased basal GR protein levels in both ShUVECs and ShUAECs. Increased GR protein levels did not result in increased dexamethasone sensitivity in the regulation of key endothelial gene expression such as endothelial nitric oxide synthase, plasminogen activator inhibitor 1, vascular endothelial growth factor, or intercellular adhesion molecule 1. In ShUVECs, MO increased GRE-dependent transactivation and FKBP prolyl isomerase 5 (FKBP5) expression. ShUAECs showed generalized glucocorticoid resistance in both dietary groups. Finally, we found that ShUVECs were less sensitive to dexamethasone-induced activation of GR than human umbilical vein endothelial cells (HUVECs). These findings suggest that MO-mediated effects in the offspring endothelium could be further mediated by dysregulation of GR homeostasis in humans as compared with sheep.
Collapse
Affiliation(s)
- Eugenia Mata-Greenwood
- Lawrence D. Longo Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.L.C.); (N.A.O.A.); (L.D.S.)
| | - Wendy L. Chow
- Lawrence D. Longo Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.L.C.); (N.A.O.A.); (L.D.S.)
| | - Nana A. O. Anti
- Lawrence D. Longo Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.L.C.); (N.A.O.A.); (L.D.S.)
| | - LeeAnna D. Sands
- Lawrence D. Longo Center for Perinatal Biology, School of Medicine, Loma Linda University, Loma Linda, CA 92350, USA; (W.L.C.); (N.A.O.A.); (L.D.S.)
| | - Olayemi Adeoye
- Department of Pharmaceutical Sciences, School of Pharmacy, Loma Linda University, Loma Linda, CA 92350, USA;
| | - Stephen P. Ford
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, WY 82071, USA (P.W.N.)
| | - Peter W. Nathanielsz
- Center for the Study of Fetal Programming, Department of Animal Science, University of Wyoming, Laramie, WY 82071, USA (P.W.N.)
| |
Collapse
|
9
|
de Carvalho VM, Chung-Filho AA, Braga FHP, Chagas-Neto P, Soares-Lima SC, Pombo-de-Oliveira MS. Interaction between birth characteristics and CRHR1, MC2R, NR3C1, GLCCI1 variants in the childhood lymphoblastic leukemia risk. Front Oncol 2024; 13:1274131. [PMID: 38348123 PMCID: PMC10859751 DOI: 10.3389/fonc.2023.1274131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/31/2023] [Indexed: 02/15/2024] Open
Abstract
Background The incidence rate of childhood acute lymphoblastic leukemia (ALL) differs worldwide, and the interplay between hemostasis actors and the maladaptive responses to environmental exposures has been explored. It has been proposed that endogenous cortisol, induced by different triggers, would eliminate pre-leukemic clones originated in utero. Herein, we tested if the interaction between CRHR1rs242941 C>A, MC2Rrs1893219 A>G, NR3C1rs41423247 G>C, and GLCCI1rs37972 C>T (players in glucocorticoid secretion) and birth characteristics would be associated with ALL risk. Methods Children aged <10 years were enrolled within the EMiLI project (period: 2012 to 2020). The study had three steps: (1) observational analysis of birth characteristics (n = 533 cases and 1,603 controls); (2) genotyping to identify single-nucleotide variants (n = 756 cases and 431 controls); and (3) case-only to test gene-environment interactions (n = 402 cases). Genetic syndromes were exclusion criteria. The controls were healthy children. The distribution of the variables was assessed through Pearson's chi-square test. Logistic regression (LR) tests were run fitted and adjusted for selected covariate models to estimate the association risk. Formal interaction analysis was also performed. Genotyping was tested by qPCR with TaqMan probes (NR3C1) or by high-resolution melting (MC2R and GLCCI1). Hardy-Weinberg equilibrium (HWE) was accessed by the chi-square test. The genotype-risk association was tested in co-dominant, dominant, and recessive models. The gene-environment interaction odds ratio (iOR) was assessed in case-only. Results Low birthweight, C-section, and low maternal schooling were associated with increased risk for ALL, adjOR 2.11, 95% CI, 1.02-4.33; adjOR 1.59, 95% CI, 1.16-2.17; and adjOR 3.78, 95% CI, 2.47-5.83, respectively, in a multiple logistic regression model. MC2R rs1893219 A>G was negatively associated with ALL (AG: OR = 0.68; 95% CI = 0.50-0.94 and GG: OR = 0.60; 95% CI = 0.42-0.85), while for GLCCI1 rs37972 C>T, TT was positively associated with ALL (OR = 1.91; 95% CI = 1.21-3.00). The combination of genotypes for MC2R (AA) and GLCCI1 (TT) increased ALL risk (OR = 2.61; 95% CI = 1.16-5.87). In a multiplicative interaction, MC2R rs1893219 A>G was associated with children whose mothers had less than 9 years of schooling (iOR = 1.99; 95% CI = 1.11-1.55). Conclusion Our study has demonstrated a significant association between MC2R rs1893219 A>G (reduced risk) and GLCCI1 rs37972 C>T variants (increased risk) and childhood ALL susceptibility. Based on this evidence, genes controlling the HPA axis activity may play a role in leukemogenesis, and further investigation is needed to substantiate our findings.
Collapse
Affiliation(s)
- Vitoria Müller de Carvalho
- Research Center, Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Alython Araujo Chung-Filho
- Research Center, Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Flávio Henrique Paraguassu Braga
- National Placental and Umbilical Cord Blood Bank, Instituto Nacional de Câncer (INCA), Ministério da Saúde (MS), Rio de Janeiro, Brazil
| | - Paulo Chagas-Neto
- Research Center, Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Sheila Coelho Soares-Lima
- Research Center, Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Maria S. Pombo-de-Oliveira
- Research Center, Molecular Carcinogenesis Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| |
Collapse
|
10
|
He W, Cui Y, Yang H, Gao J, Zhao Y, Hao N, Li Y, Zhang M. Aquatic toxicity, ecological effects, human exposure pathways and health risk assessment of liquid crystal monomers. JOURNAL OF HAZARDOUS MATERIALS 2024; 461:132681. [PMID: 37801980 DOI: 10.1016/j.jhazmat.2023.132681] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/19/2023] [Accepted: 09/29/2023] [Indexed: 10/08/2023]
Abstract
Liquid crystal monomers (LCMs), one of the key materials for liquid crystal displays, have been considered as emerging pollutants in recent years. However, the environmental behaviors of LCMs have not yet been well investigated. The toxicity data of 1173 LCMs were calculated by integrated computational simulation methods in this study. It showed that 64.6% LCMs exhibited PBT (persistent, bioaccumulative, and toxic) properties. Based on the results, 1173 LCMs were identified as molecules possessing the highest level of acute toxicity to aquatic organisms. Among which, and a human health risk priority control list about LCMs was generated in this study, among which 435 were classified as requiring priority control LCMs. It was confirmed that LCMs could eventually accumulate in the human body along the aquatic food chain or penetrate the bloodstream through the dermis, thereby causing harm to health by identifying the exposure pathways of LCMs in humans. Additionally, the electronegativity of the side chain group of LCMs is the main factor causing toxicity differences; therefore, the LCMs containing halogens presented significant acute and chronic toxic effects. This study provided a more comprehensive understanding of LCMs for the public and scientific strategies for controlling LCMs.
Collapse
Affiliation(s)
- Wei He
- MOE Key Laboratory of Resources Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China
| | - Yuhan Cui
- MOE Key Laboratory of Resources Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China
| | - Hao Yang
- MOE Key Laboratory of Resources Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China
| | - Jiaxuan Gao
- MOE Key Laboratory of Resources Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China
| | - Yuanyuan Zhao
- MOE Key Laboratory of Resources Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China
| | - Ning Hao
- College of New Energy and Environment, Jilin University, Changchun 130012, China
| | - Yu Li
- MOE Key Laboratory of Resources Environmental Systems Optimization, North China Electric Power University, Beijing 102206, China
| | - Meng Zhang
- College of Environmental Sciences and Engineering, State Environmental Protection Key Laboratory of All Material Fluxes in River Ecosystems, Peking University, Beijing 100871, China; The Key Laboratory of Water and Sediment Sciences, Ministry of Education, International Joint Laboratory for Regional Pollution Control, Ministry of Education, Beijing 100871, China.
| |
Collapse
|
11
|
Halcomb CJ, Philipp TR, Dhillon PS, Cox JH, Aguilar-Alvarez R, Vanderhoof SO, Jasnow AM. Sex divergent behavioral responses in platform-mediated avoidance and glucocorticoid receptor blockade. Psychoneuroendocrinology 2024; 159:106417. [PMID: 37925931 PMCID: PMC10872426 DOI: 10.1016/j.psyneuen.2023.106417] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/17/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
Women are more likely than men to develop anxiety or stress-related disorders. A core behavioral symptom of all anxiety disorders is avoidance of fear or anxiety eliciting cues. Recent rodent models of avoidance show reliable reproduction of this behavioral phenomenon in response to learned aversive associations. Here, a modified version of platform-mediated avoidance that lacked an appetitive task was utilized to investigate the learning and extinction of avoidance in male and female C57BL6/J mice. Here, we found a robust sex difference in the acquisition and extinction of platform-mediated avoidance. Across three experiments, 63.7% of female mice acquired avoidance according to our criterion, whereas 83.8% of males acquired it successfully. Of those females that acquired avoidance, they displayed persistent avoidance after extinction compared to males. Given their role in regulating stress responses and habitual behaviors, we investigated if glucocorticoid receptors (GR) mediated avoidance learning in males and females. We found that a subcutaneous injection (25 mg/kg) of the GR antagonist, RU486 (Mifepristone), significantly reduced persistent avoidance in females but did not further reduce avoidance in males after extinction. These data suggest that GR activation during avoidance learning may contribute to persistent avoidance in females that is resistant to extinction.
Collapse
Affiliation(s)
- Carly J Halcomb
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Trey R Philipp
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Parker S Dhillon
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - J Hunter Cox
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Ricardo Aguilar-Alvarez
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | | | - Aaron M Jasnow
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29209, USA.
| |
Collapse
|
12
|
Gore IR, Gould E. Developmental and adult stress: effects of steroids and neurosteroids. Stress 2024; 27:2317856. [PMID: 38563163 PMCID: PMC11046567 DOI: 10.1080/10253890.2024.2317856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 02/03/2024] [Indexed: 04/04/2024] Open
Abstract
In humans, exposure to early life adversity has profound implications for susceptibility to developing neuropsychiatric disorders later in life. Studies in rodents have shown that stress experienced during early postnatal life can have lasting effects on brain development. Glucocorticoids and sex steroids are produced in endocrine glands and the brain from cholesterol; these molecules bind to nuclear and membrane-associated steroid receptors. Unlike other steroids that can also be made in the brain, neurosteroids bind specifically to neurotransmitter receptors, not steroid receptors. The relationships among steroids, neurosteroids, and stress are multifaceted and not yet fully understood. However, studies demonstrating altered levels of progestogens, androgens, estrogens, glucocorticoids, and their neuroactive metabolites in both developmental and adult stress paradigms strongly suggest that these molecules may be important players in stress effects on brain circuits and behavior. In this review, we discuss the influence of developmental and adult stress on various components of the brain, including neurons, glia, and perineuronal nets, with a focus on sex steroids and neurosteroids. Gaining an enhanced understanding of how early adversity impacts the intricate systems of brain steroid and neurosteroid regulation could prove instrumental in identifying novel therapeutic targets for stress-related conditions.
Collapse
Affiliation(s)
- Isha R Gore
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Elizabeth Gould
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| |
Collapse
|
13
|
Zhang Q, Xu W, Kong Z, Wu Y, Liu Y. Cadmium exposure-induced rat testicular dysfunction and its mechanism of chronic stress. Food Chem Toxicol 2023; 182:114181. [PMID: 37972751 DOI: 10.1016/j.fct.2023.114181] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/23/2023] [Accepted: 11/03/2023] [Indexed: 11/19/2023]
Abstract
Cadmium is a common environmental pollutant in daily life, the toxic mechanisms of chronic cadmium exposure on the testes have not been fully elucidated. This study aimed to explore the effects of cadmium exposure on male reproductive health and its mechanism. The results showed that cadmium exposure led widened interstitial spaces, abnormal seminiferous tubule morphology, and decreased Leydig cell numbers. Moreover, sperm quality was significantly reduced, along with a decrease in fertility rate. And cadmium exposure could activate the hypothalamic-pituitary-adrenal (HPA) axis, elevate blood glucocorticoid levels, subsequently increase glucocorticoid receptor (GR) expression and activation in testicular Leydig cells. Then GR act on the glucocorticoid receptor element (GRE) in the DNA methyltransferase 3 A (DNMT3A) promoter region and upregulate DNMT3A expression. Consequently, this led to an increase in DNA methylation levels in the angiotensin II receptor 2 (AT2R) promoter region, resulting in reduced AT2R expression and inhibiting testicular steroidogenesis. This study systematically elucidated that cadmium exposure could lead to testicular steroidogenesis suppression and decreased fertility through the GR/DNMT3A/AT2R signaling pathway. This research further provides theoretical and experimental evidence for confirming the threat of cadmium exposure to human reproduction, and contributes to the guidance and protection of male reproductive health.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China
| | - Wei Xu
- Department of Neurology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China
| | - ZiYu Kong
- Department of Pharmacology, TaiKang Medical School (School of Basic Medical Sciences), Wuhan University, Wuhan, 430071, China
| | - YuJiao Wu
- Department of Clinical Pharmacy, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, Hubei, 445000, China.
| | - Yi Liu
- China Tobacco HuBei Industrial LLC, Wuhan, 430071, China.
| |
Collapse
|
14
|
Stojceski F, Buetti-Dinh A, Stoddart MJ, Danani A, Della Bella E, Grasso G. Influence of dexamethasone on the interaction between glucocorticoid receptor and SOX9: A molecular dynamics study. J Mol Graph Model 2023; 125:108587. [PMID: 37579519 DOI: 10.1016/j.jmgm.2023.108587] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/16/2023]
Abstract
The glucocorticoid receptor (GR) is a nuclear receptor that controls critical biological processes by regulating the transcription of specific genes. GR transcriptional activity is modulated by a series of ligands and coenzymes, where a ligand can act as an agonist or antagonist. GR agonists, such as the glucocorticoids dexamethasone (DEX) and prednisolone, are widely prescribed to patients with inflammatory and autoimmune diseases. DEX is also used to induce osteogenic differentiation in vitro. Recently, it has been highlighted that DEX induces changes in the osteogenic differentiation of human mesenchymal stromal cells by downregulating the transcription factor SRY-box transcription factor 9 (SOX9) and upregulating the peroxisome proliferator-activated receptor γ (PPARG). SOX9 is fundamental in the control of chondrogenesis, but also in osteogenesis by acting as a dominant-negative of RUNX2. Many processes remain to be clarified during cell fate determination, such as the interplay between the key transcription factors. The main objective pursued by this work is to shed light on the interaction between GR and SOX9 in the presence and absence of DEX at an atomic level of resolution using molecular dynamics simulations. The outcome of this research could help the understanding of possible molecular interactions between GR and SOX9 and their role in the determination of cell fate. The results highlight the key residues at the interface between GR and SOX9 involved in the complexation process and shed light on the mechanism through which DEX modulates GR-SOX9 binding and exerts its biological activity.
Collapse
Affiliation(s)
- Filip Stojceski
- Dalle Molle Institute for Artificial Intelligence USI-SUPSI Polo universitario Lugano - Campus Est, Via la Santa 1, 6962, Lugano-Viganello, Switzerland
| | - Antoine Buetti-Dinh
- Dalle Molle Institute for Artificial Intelligence USI-SUPSI Polo universitario Lugano - Campus Est, Via la Santa 1, 6962, Lugano-Viganello, Switzerland
| | - Martin J Stoddart
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland
| | - Andrea Danani
- Dalle Molle Institute for Artificial Intelligence USI-SUPSI Polo universitario Lugano - Campus Est, Via la Santa 1, 6962, Lugano-Viganello, Switzerland
| | - Elena Della Bella
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland.
| | - Gianvito Grasso
- Dalle Molle Institute for Artificial Intelligence USI-SUPSI Polo universitario Lugano - Campus Est, Via la Santa 1, 6962, Lugano-Viganello, Switzerland.
| |
Collapse
|
15
|
Zou H, Yu J, Li Z, Liu Y, Wang T, Li T, Lv C, Zhang J. In vitro, in vivo, and in silico evaluation of the glucocorticoid receptor antagonist activity of 3,6-dibromocarbazole. Food Chem Toxicol 2023; 180:114048. [PMID: 37734465 DOI: 10.1016/j.fct.2023.114048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
3,6-Dibromocarbazole is a novel environmental contaminant which is currently detected in several environmental media worldwide. This work aims to investigate the anti-glucocorticoid potency and endocrine disrupting effects of 3,6-dibromocarbazole. In vitro experiments indicated that 3,6-dibromocarbazole possessed glucocorticoid receptor (GR) antagonistic activity and inhibited dexamethasone-induced GR nuclear translocation. 3,6-Dibromocarbazole reduced the expression levels of glucocorticoid responsive genes including glucose-6-phosphatase (G6Pase), phosphoenolpyruvate carboxykinase (PEPCK), fatty acid synthase (FAS), and tyrosine aminotransferase (TAT), and further disrupted the protein expression of two key enzymes PEPCK and FAS in gluconeogenesis. In vivo experiments showed that 3,6-dibromocarbazole induced abnormal development of zebrafish embryos and disrupted the major neurohormones involved in activation of hypothalamic-pituitary-adrenocortical (HPA) axis in zebrafish larvae. The results of molecular docking and molecular dynamics simulation contributed to explain the antagonistic effect of 3,6-dibromocarbazole. Taken together, this work identified 3,6-dibromocarbazole as a GR antagonist, which might exert endocrine disrupting effects by interfering the pathway of gluconeogenesis.
Collapse
Affiliation(s)
- Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Jia Yu
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Zhuolin Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Yao Liu
- College of Food and Bioengineering, Qiqihar University, Qiqihar, 161006, China
| | - Tuoyi Wang
- College of Food and Bioengineering, Qiqihar University, Qiqihar, 161006, China
| | - Tiezhu Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Chengyu Lv
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| |
Collapse
|
16
|
Halcomb CJ, Philipp TR, Dhillon PS, Cox JH, Aguilar-Alvarez R, Vanderhoof SO, Jasnow AM. Sex divergent behavioral responses in platform-mediated avoidance and glucocorticoid receptor blockade. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.26.559122. [PMID: 37808636 PMCID: PMC10557728 DOI: 10.1101/2023.09.26.559122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Women are more likely than men to develop anxiety or stress-related disorders. A core behavioral symptom of all anxiety disorders is avoidance of fear or anxiety eliciting cues. Recent rodent models of avoidance show reliable reproduction of this behavioral phenomenon in response to learned aversive associations. Here, a modified version of platform-mediated avoidance that lacked an appetitive task was utilized to investigate the learning and extinction of avoidance in male and female C57BL6/J mice. Here, we found a robust sex difference in the acquisition and extinction of platform-mediated avoidance. Across three experiments, 63.7% of female mice acquired avoidance according to our criterion, whereas 83.8% of males acquired it successfully. Of those females that acquired avoidance, they displayed persistent avoidance after extinction compared to males. Given their role in regulating stress responses and habitual behaviors, we investigated if glucocorticoid receptors (GR) mediated avoidance learning in males and females. Here we found that a subcutaneous injection (25mg/kg) of the GR antagonist, RU486 (mifepristone), significantly reduced persistent avoidance in females but did not further reduce avoidance in males after extinction. These data suggest that GR activation during avoidance learning may contribute to persistent avoidance in females that is resistant to extinction.
Collapse
|
17
|
Zafar A, Khan MJ, Naeem A. MDM2- an indispensable player in tumorigenesis. Mol Biol Rep 2023; 50:6871-6883. [PMID: 37314603 PMCID: PMC10374471 DOI: 10.1007/s11033-023-08512-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/10/2023] [Indexed: 06/15/2023]
Abstract
Murine double minute 2 (MDM2) is a well-recognized molecule for its oncogenic potential. Since its identification, various cancer-promoting roles of MDM2 such as growth stimulation, sustained angiogenesis, metabolic reprogramming, apoptosis evasion, metastasis, and immunosuppression have been established. Alterations in the expression levels of MDM2 occur in multiple types of cancers resulting in uncontrolled proliferation. The cellular processes are modulated by MDM2 through transcription, post-translational modifications, protein degradation, binding to cofactors, and subcellular localization. In this review, we discuss the precise role of deregulated MDM2 levels in modulating cellular functions to promote cancer growth. Moreover, we also briefly discuss the role of MDM2 in inducing resistance against anti-cancerous therapies thus limiting the benefits of cancerous treatment.
Collapse
Affiliation(s)
- Aasma Zafar
- Department of Biosciences, COMSATS University, Islamabad, 45550 Pakistan
| | | | - Aisha Naeem
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, 20057 Washington, DC U.S
- Qatar University Health, Qatar University, P.O. Box 2713, Doha, Qatar
| |
Collapse
|
18
|
Hu Y, Li J, Li X, Wang D, Xiang R, Liu W, Hou S, Zhao Q, Yu X, Xu M, Zhao D, Li T, Chi Y, Yang J. Hepatocyte-secreted FAM3D ameliorates hepatic steatosis by activating FPR1-hnRNP U-GR-SCAD pathway to enhance lipid oxidation. Metabolism 2023:155661. [PMID: 37454871 DOI: 10.1016/j.metabol.2023.155661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide; however, the underlying mechanisms remain poorly understood. FAM3D is a member of the FAM3 family; however, its role in hepatic glycolipid metabolism remains unknown. Serum FAM3D levels are positively correlated with fasting blood glucose levels in patients with diabetes. Hepatocytes express and secrete FAM3D, and its expression is increased in steatotic human and mouse livers. Hepatic FAM3D overexpression ameliorated hyperglycemia and steatosis in obese mice, whereas FAM3D-deficient mice exhibited exaggerated hyperglycemia and steatosis after high-fat diet (HFD)-feeding. In cultured hepatocytes, FAM3D overexpression or recombinant FAM3D protein (rFAM3D) treatment reduced gluconeogenesis and lipid deposition, which were blocked by anti-FAM3D antibodies or inhibition of its receptor, formyl peptide receptor 1 (FPR1). FPR1 overexpression suppressed gluconeogenesis and reduced lipid deposition in wild hepatocytes but not in FAM3D-deficient hepatocytes. The addition of rFAM3D restored FPR1's inhibitory effects on gluconeogenesis and lipid deposition in FAM3D-deficient hepatocytes. Hepatic FPR1 overexpression ameliorated hyperglycemia and steatosis in obese mice. RNA sequencing and DNA pull-down revealed that the FAM3D-FPR1 axis upregulated the expression of heterogeneous nuclear ribonucleoprotein U (hnRNP U), which recruits the glucocorticoid receptor (GR) to the promoter region of the short-chain acyl-CoA dehydrogenase (SCAD) gene, promoting its transcription to enhance lipid oxidation. Moreover, FAM3D-FPR1 axis also activates calmodulin-Akt pathway to suppress gluconeogenesis in hepatocytes. In conclusion, hepatocyte-secreted FAM3D activated the FPR1-hnRNP U-GR-SCAD pathway to enhance lipid oxidation in hepatocytes. Under obesity conditions, increased hepatic FAM3D expression is a compensatory mechanism against dysregulated glucose and lipid metabolism.
Collapse
Affiliation(s)
- Yuntao Hu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Jing Li
- Department of Endocrinology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100027, China
| | - Xin Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Di Wang
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China
| | - Rui Xiang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Wenjun Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Song Hou
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Qinghe Zhao
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
| | - Xiaoxing Yu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China
| | - Ming Xu
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Science of the Ministry of Education, Beijing 100191, China
| | - Dong Zhao
- Department of Endocrinology, Beijing Luhe Hospital, Capital Medical University, Beijing 101100, China
| | - Tao Li
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing 100044, China
| | - Yujing Chi
- Department of Central Laboratory and Institute of Clinical Molecular Biology, Peking University People's Hospital, Beijing 100044, China; Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China.
| | - Jichun Yang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Center for Non-coding RNA Medicine, Peking University Health Science Center, Beijing 100191, China; Department of Cardiology, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
19
|
Choi D, Kang W, Park S, Son B, Park T. Identification of Glucocorticoid Receptor Target Genes That Potentially Inhibit Collagen Synthesis in Human Dermal Fibroblasts. Biomolecules 2023; 13:978. [PMID: 37371558 DOI: 10.3390/biom13060978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Over several decades, excess glucocorticoids (GCs) of endogenous or exogenous origin have been recognized to significantly inhibit collagen synthesis and accelerate skin aging. However, little is known regarding their molecular mechanisms. We hypothesized that the action of GCs on collagen production is at least partially through the glucocorticoid receptor (GR) and its target genes, and therefore aimed to identify GR target genes that potentially inhibit collagen synthesis in Hs68 human dermal fibroblasts. We first confirmed that dexamethasone, a synthetic GC, induced canonical GR signaling in dermal fibroblasts. We then collected 108 candidates for GR target genes reported in previous studies on GR target genes and verified that 17 genes were transcriptionally upregulated in dexamethasone-treated dermal fibroblasts. Subsequently, by individual knockdown of the 17 genes, we identified that six genes, AT-rich interaction domain 5B, FK506 binding protein 5, lysyl oxidase, methylenetetrahydrofolate dehydrogenase (NADP + dependent) 2, zinc finger protein 36, and zinc fingers and homeoboxes 3, are potentially involved in GC-mediated inhibition of collagen synthesis. The present study sheds light on the molecular mechanisms of GC-mediated skin aging and provides a basis for further research on the biological characteristics of individual GR target genes.
Collapse
Affiliation(s)
- Dabin Choi
- Department of Food and Nutrition, BK21 FOUR, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Republic of Korea
| | - Wesuk Kang
- Department of Food and Nutrition, BK21 FOUR, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Republic of Korea
| | - Soyoon Park
- Department of Food and Nutrition, BK21 FOUR, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Republic of Korea
| | - Bomin Son
- Department of Food and Nutrition, BK21 FOUR, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Republic of Korea
| | - Taesun Park
- Department of Food and Nutrition, BK21 FOUR, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 120-749, Republic of Korea
| |
Collapse
|
20
|
NR3C1 overexpression regulates the expression and alternative splicing of inflammation-associated genes involved in PTSD. Gene 2023; 859:147199. [PMID: 36657650 DOI: 10.1016/j.gene.2023.147199] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/13/2022] [Accepted: 01/10/2023] [Indexed: 01/18/2023]
Abstract
NR3C1-encoding glucocorticoid receptors have dual roles as RNA-binding protein and transcription factor. Recent studies revealed that NR3C1 might play an important role in the pathogenesis of PTSD (Post-traumatic stress disorder). However, its molecular mechanism remained unclear. In the present study, a neuronal cell model was constructed by transfecting a NR3C1-overexpressing plasmid pIRES-hrGFP-1a-NR3C1 or empty vector into HT22 cells. The changes in global transcription levels and alternative splicing events in HT22 cells after NR3C1 overexpression were analyzed by RNA sequencing. Compared with the empty vector control, the expression of inflammatory factors were differentially regulated by NR3C1, including genes involved in chemokine signal pathway, PI3K-Akt signal pathway, cytokine receptor interaction, neural ligand-receptor interaction and so on. In addition, NR3C1 regulated the alternative splicing of many genes involved in immune response, axon formation, stress response and inflammation. This study was the first to perform a transcriptome analysis of differential gene expression and alternative splicing in a NR3C1-overexpressing HT22 cell model. Our results suggested that NR3C1 could manipulate the expression of inflammatory transcription factors and their alternative splicing patterns, subsequently affecting the expression of downstream targets, may be leading to the onset of PTSD. This study will provide new insights in the NR3C1-mediated gene regulation in relation to PTSD.
Collapse
|
21
|
Jeon H, Choi Y, Brännström M, Akin JW, Curry TE, Jo M. Cortisol/glucocorticoid receptor: a critical mediator of the ovulatory process and luteinization in human periovulatory follicles. Hum Reprod 2023; 38:671-685. [PMID: 36752644 PMCID: PMC10068287 DOI: 10.1093/humrep/dead017] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 01/03/2023] [Indexed: 02/09/2023] Open
Abstract
STUDY QUESTION Do cortisol/glucocorticoid receptors play an active role in the human ovary during ovulation and early luteinization? SUMMARY ANSWER The ovulatory hCG stimulation-induced glucocorticoid receptor signaling plays a crucial role in regulating steroidogenesis and ovulatory cascade in human periovulatory follicles. WHAT IS KNOWN ALREADY Previous studies reported an increase in cortisol levels in the human follicular fluid after the LH surge or ovulatory hCG administration. However, little is known about the role of cortisol/glucocorticoid receptors in the ovulatory process and luteinization in humans. STUDY DESIGN, SIZE, DURATION This study was an experimental prospective clinical and laboratory-based study. An in vivo experimental study was accomplished utilizing the dominant ovarian follicles from 38 premenopausal women undergoing laparoscopic sterilization. An in vitro experimental study was completed using the primary human granulosa/lutein cells (hGLC) from 26 premenopausal women undergoing IVF. PARTICIPANTS/MATERIALS, SETTING, METHODS This study was conducted in a private fertility clinic and academic medical centers. Dominant ovarian follicles were collected before the LH surge and at defined times after hCG administration from women undergoing laparoscopic sterilization. Primary hGLC were collected from women undergoing IVF. hGLC were treated without or with hCG in the absence or presence of RU486 (20 µM; dual antagonist for progesterone receptor and glucocorticoid receptor) or CORT125281 (50 µM; selective glucocorticoid receptor antagonist) for 12 or 36 h. The expression of genes involved in glucocorticoid receptor signaling, steroidogenesis, and ovulatory cascade was studied with RT-quantitative PCR and western blotting. The production of cortisol, corticosterone, and progesterone was assessed by hormone assay kits. MAIN RESULTS AND THE ROLE OF CHANCE hCG administration upregulated the expression of hydroxysteroid 11-beta dehydrogenase 1 (HSD11B1), nuclear receptor subfamily 3 group C member 1 (NR3C1), FKBP prolyl isomerase 5 (FKBP5), and FKBP prolyl isomerase 4 (FKBP4) in human ovulatory follicles and in hGLC (P < 0.05). RU486 and CORT125281 reduced hCG-induced increases in progesterone and cortisol production in hGLC. The expression of genes involved in glucocorticoid receptor signaling, steroidogenesis, and the key ovulatory process was reduced by RU486 and/or CORT125281 in hGLC. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The role of cortisol/glucocorticoid receptors demonstrated using the hGLC model may not fully reflect their physiological roles in vivo. WIDER IMPLICATIONS OF THE FINDINGS Successful ovulation and luteinization are essential for female fertility. Women with dysregulated cortisol levels often suffer from anovulatory infertility. Deciphering the functional role of glucocorticoid receptor signaling in human periovulatory follicles enhances our knowledge of basic ovarian physiology and may provide therapeutic insights into treating infertility in women. STUDY FUNDING/COMPETING INTEREST(S) This study was supported by P01HD71875 (to M.J., T.E.C., and M.B.) and R01HD096077 (to M.J.) from the Foundation for the National Institutes of Health and the BTPSRF of the University of Kentucky Markey Cancer Center (P30CA177558). The authors report no competing interests. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- H Jeon
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, USA
| | - Y Choi
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - M Brännström
- Department of Obstetrics and Gynecology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Stockholm IVF, Stockholm, Sweden
| | - J W Akin
- Bluegrass Fertility Center, Lexington, KY, USA
| | - T E Curry
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| | - M Jo
- Department of Obstetrics and Gynecology, University of Kentucky College of Medicine, Lexington, KY, USA
| |
Collapse
|
22
|
CTNNAL1 enhances glucocorticoid sensitivity in HDM-induced asthma mouse model through deactivating hsp90 signaling pathway. Life Sci 2023; 313:121304. [PMID: 36535402 DOI: 10.1016/j.lfs.2022.121304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
AIMS Adhesion molecules play vital roles in the induction of airway hyperresponsiveness (AHR) or airway inflammation. The down-regulation of catenin alpha-like 1 (CTNNAL1) in the bronchial epithelial cells of asthma patients and mice models has been noted in our previous study. In this work, we further explore the underlying mechanism of CTNNAL1 in asthma. MAIN METHODS We constructed a house dust mite (HDM)-induced asthma animal model on control mice and applied CTNNAL1-siRNA transfection to create CTNNAL1-deficient mice. KEY FINDINGS We documented much more severe airway inflammation and increased leukocyte infiltration in the lungs of the CTNNAL1-deficient mice comparing to control mice, along with elevated expression of inflammatory cytokines. Dexamethasone (DEX) treatment led to less reduced inflammation in CTNNAL1-deficient mice compared with control mice. Immunoprecipitation confirmed the interaction between heat shock protein90 (hsp90) and CTNNAL1. The expression of hsp90 was upregulated after CTNNAL1 silencing. Meanwhile, the use of hsp90 inhibitor geldanamycin significantly decreased the expression of NR3C1, ICAM-1 and the ratio of p-p65/p65 in CTNNAL1-silenced 16HBE14o- cells. Both geldanamycin and DEX could function to suppress the expression of ICAM-1 and the phosphorylation level of p65. Nevertheless, the anti-inflammatory effect of DEX proved less potent than geldanamycin in the CTNNAL1-silenced group. The combined therapy of geldanamycin and DEX significantly decreased the inflammatory responses in CTNNAL1-deficient HBE cells than DEX monotherapy. SIGNIFICANCE Our study corroborates that CTNNAL1 deficiency induced aggravated airway inflammation and rendered insensitivity to glucocorticoids via triggering hsp90 signaling pathway.
Collapse
|
23
|
Mitsis T, Papageorgiou L, Papakonstantinou E, Diakou I, Pierouli K, Dragoumani K, Bacopoulou F, Kino T, Chrousos GP, Eliopoulos E, Vlachakis D. A Genomic Study of the Japanese Population Focusing on the Glucocorticoid Receptor Interactome Highlights Distinct Genetic Characteristics Associated with Stress Response. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1423:101-113. [PMID: 37525035 DOI: 10.1007/978-3-031-31978-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
All living organisms have been programmed to maintain a complex inner equilibrium called homeostasis, despite numerous adversities during their lifespan. Any threatening or perceived as such stimuli for homeostasis is termed a stressor, and a highly conserved response system called the stress response system has been developed to cope with these stimuli and maintain or reinstate homeostasis. The glucocorticoid receptor, a transcription factor belonging to the nuclear receptors protein superfamily, has a major role in the stress response system, and research on its interactome may provide novel information regarding the mechanisms underlying homeostasis maintenance. A list of 149 autosomal genes that have an essential role in GR function or are prime examples of GRE-containing genes was composed in order to gain a comprehensive view of the GR interactome. A search for SNPs on those particular genes was conducted on a dataset of 3554 Japanese individuals, with mentioned polymorphisms being annotated with relevant information from the ClinVar, LitVar, and dbSNP databases. Forty-two SNPs of interest and their genomic locations were identified. These SNPs have been associated with drug metabolism and neuropsychiatric, metabolic, and immune system disorders, while most of them were located in intronic regions. The frequencies of those SNPs were later compared with a dataset consisting of 1465 Korean individuals in order to find population-specific characteristics based on some of the identified SNPs of interest. The results highlighted.that rs1043618 frequencies were different in the two populations, with mentioned polymorphism having a potential role in chronic obstructive pulmonary disease in response to environmental stressors. This SNP is located in the HSPA1A gene, which codes for an essential GR co-chaperone, and such information showcases that similar gene may be novel genomic targets for managing or combatting stress-related pathologies.
Collapse
Affiliation(s)
- Thanasis Mitsis
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Louis Papageorgiou
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Eleni Papakonstantinou
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Io Diakou
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Katerina Pierouli
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Konstantina Dragoumani
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Flora Bacopoulou
- University Research Institute of Maternal and Child Health & Precision Medicine, Athens, Greece
- National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Tomoshige Kino
- Department of Human Genetics, Division of Translational Medicine, Sidra Medical and Research Center, Doha, Qatar
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, Athens, Greece
- National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Elias Eliopoulos
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Dimitrios Vlachakis
- Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece.
- University Research Institute of Maternal and Child Health & Precision Medicine, Athens, Greece.
- National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece.
- Division of Endocrinology and Metabolism, Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
24
|
Tylan C, Engler HI, Villar G, Langkilde T. Consumption of fire ants, an invasive predator and prey of native lizards, may enhance immune functions needed to combat envenomation. Biol Invasions 2022. [DOI: 10.1007/s10530-022-02939-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
25
|
Dexamethasone-Induced Adipose Tissue Redistribution and Metabolic Changes: Is Gene Expression the Main Factor? An Animal Model of Chronic Hypercortisolism. Biomedicines 2022; 10:biomedicines10092328. [PMID: 36140428 PMCID: PMC9496558 DOI: 10.3390/biomedicines10092328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/03/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Chronic hypercortisolism has been associated with the development of several metabolic alterations, mostly caused by the effects of chronic glucocorticoid (GC) exposure over gene expression. The metabolic changes can be partially explained by the GC actions on different adipose tissues (ATs), leading to central obesity. In this regard, we aimed to characterize an experimental model of iatrogenic hypercortisolism in rats with significant AT redistribution. Male Wistar rats were distributed into control (CT) and GC-treated, which received dexamethasone sodium phosphate (0.5 mg/kg/day) by an osmotic minipump, for 4 weeks. GC-treated rats reproduced several characteristics observed in human hypercortisolism/Cushing’s syndrome, such as HPA axis inhibition, glucose intolerance, insulin resistance, dyslipidemia, hepatic lipid accumulation, and AT redistribution. There was an increase in the mesenteric (meWAT), perirenal (prWAT), and interscapular brown (BAT) ATs mass, but a reduction of the retroperitoneal (rpWAT) mass compared to CT rats. Overexpressed lipolytic and lipogenic gene profiles were observed in white adipose tissue (WAT) of GC rats as BAT dysfunction and whitening. The AT remodeling in response to GC excess showed more importance than the increase of AT mass per se, and it cannot be explained just by GC regulation of gene transcription.
Collapse
|
26
|
Li JX, Cummins CL. Fresh insights into glucocorticoid-induced diabetes mellitus and new therapeutic directions. Nat Rev Endocrinol 2022; 18:540-557. [PMID: 35585199 PMCID: PMC9116713 DOI: 10.1038/s41574-022-00683-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 02/08/2023]
Abstract
Glucocorticoid hormones were discovered to have use as potent anti-inflammatory and immunosuppressive therapeutics in the 1940s and their continued use and development have successfully revolutionized the management of acute and chronic inflammatory diseases. However, long-term use of glucocorticoids is severely hampered by undesirable metabolic complications, including the development of type 2 diabetes mellitus. These effects occur due to glucocorticoid receptor activation within multiple tissues, which results in inter-organ crosstalk that increases hepatic glucose production and inhibits peripheral glucose uptake. Despite the high prevalence of glucocorticoid-induced hyperglycaemia associated with their routine clinical use, treatment protocols for optimal management of the metabolic adverse effects are lacking or underutilized. The type, dose and potency of the glucocorticoid administered dictates the choice of hypoglycaemic intervention (non-insulin or insulin therapy) that should be provided to patients. The longstanding quest to identify dissociated glucocorticoid receptor agonists to separate the hyperglycaemic complications of glucocorticoids from their therapeutically beneficial anti-inflammatory effects is ongoing, with selective glucocorticoid receptor modulators in clinical testing. Promising areas of preclinical research include new mechanisms to disrupt glucocorticoid signalling in a tissue-selective manner and the identification of novel targets that can selectively dissociate the effects of glucocorticoids. These research arms share the ultimate goal of achieving the anti-inflammatory actions of glucocorticoids without the metabolic consequences.
Collapse
Affiliation(s)
- Jia-Xu Li
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada.
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
27
|
Yao YZ, Brennan FE, Carvajal CA, Vecchiola A, Tapia-Castillo A, Fardella CE, Fuller PJ. Cortisol resistance in the degu (Octodon degus). Steroids 2022; 184:109037. [PMID: 35429494 DOI: 10.1016/j.steroids.2022.109037] [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: 01/28/2022] [Revised: 03/23/2022] [Accepted: 04/11/2022] [Indexed: 11/21/2022]
Abstract
Cortisol resistance has also been reported in the degu, Octodon degus, a New World hystricomorph endemic to central Chile. The degu is used as a model for studies of stress and diurnal rhythms, parental behaviour and female masculinization. Another New World hystricomorph, the guinea pig, also exhibits glucocorticoid resistance, a result of amino acid sequences that differ from other mammalian glucocorticoid receptors (GR). Mutations in the ligand-binding domain of the human GR have been identified in familial or sporadic generalised cortisol resistance as have variants in the guinea pig. To address the possibility that the high levels of cortisol observed in the degu are a result of the same or similar sequence variations observed in the guinea pig GR, we have cloned, expressed and characterised the ligand-binding domain (LBD) of the degu GR. Somewhat unexpectedly, neither the amino acids nor the region involved in the resistance observed in the guinea pig GR are relevant in the degu GR. The relative resistance to cortisol observed in the degu GR is conferred by the substitution of two isoleucine residues, which are highly conserved in the GR across species, with a valine doublet. These amino acids lie in the region between helices 5 and 6 of the GR LBD, a region known to be important in determining the affinity of ligand-binding in steroid receptors.
Collapse
Affiliation(s)
- Yi-Zhou Yao
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Monash University, Department of Molecular Translational Science, Clayton, Victoria 3168, Australia
| | - Francine E Brennan
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Monash University, Department of Molecular Translational Science, Clayton, Victoria 3168, Australia
| | - Cristian A Carvajal
- Departamento de Endocrinologia, Facultad de Medicina, and Centro Traslacional de Endocrinología (CETREN UC) Pontificia, Universidad Catolica de Chile, Santiago, Chile
| | - Andrea Vecchiola
- Departamento de Endocrinologia, Facultad de Medicina, and Centro Traslacional de Endocrinología (CETREN UC) Pontificia, Universidad Catolica de Chile, Santiago, Chile
| | - Alejandra Tapia-Castillo
- Departamento de Endocrinologia, Facultad de Medicina, and Centro Traslacional de Endocrinología (CETREN UC) Pontificia, Universidad Catolica de Chile, Santiago, Chile
| | - Carlos E Fardella
- Departamento de Endocrinologia, Facultad de Medicina, and Centro Traslacional de Endocrinología (CETREN UC) Pontificia, Universidad Catolica de Chile, Santiago, Chile
| | - Peter J Fuller
- Centre for Endocrinology and Metabolism, Hudson Institute of Medical Research and the Monash University, Department of Molecular Translational Science, Clayton, Victoria 3168, Australia.
| |
Collapse
|
28
|
Duan P, Wang H, Yi X, Zhang H, Chen H, Pan Z. C/EBPα regulates the fate of bone marrow mesenchymal stem cells and steroid-induced avascular necrosis of the femoral head by targeting the PPARγ signalling pathway. Stem Cell Res Ther 2022; 13:342. [PMID: 35883192 PMCID: PMC9327281 DOI: 10.1186/s13287-022-03027-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 07/02/2022] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND The imbalance of osteogenic/adipogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) is closely related to steroid-induced avascular necrosis of the femoral head (SANFH). We aimed to investigate the epigenetic mechanism of intramedullary fat accumulation and continuous osteonecrosis after glucocorticoid (GC) withdrawal in SANFH. METHODS An SANFH model was established in SD rats, which received an intermittent high GC dose for the first 4 weeks followed by an additional 4 weeks without GC. We explored the synergistic effects and mechanisms of C/EBPα and PPARγ on the differentiation of BMSCs by lentivirus-mediated gene knockdown and overexpression assays. A chromatin immunoprecipitation assay was performed to identify epigenetic modification sites on PPARγ in vivo and in vitro. RESULTS In the SANFH model, intramedullary fat was significantly increased, and the transcription factors C/EBPα and PPARγ were upregulated simultaneously in the femoral head. In vitro, C/EBPα promoted adipogenic differentiation of BMSCs by targeting the PPARγ signalling pathway, while overexpression of C/EBPα significantly impaired osteogenic differentiation. Further studies demonstrated that histone H3K27 acetylation of PPARγ played an important role in the epigenetic mechanism underlying SANFH. C/EBPα upregulates the histone H3K27 acetylation level in the PPARγ promoter region by inhibiting HDAC1. Additionally, inhibiting the histone acetylation level of PPARγ effectively prevented adipogenic differentiation, thus slowing the progression of SANFH. CONCLUSIONS Our results demonstrate the molecular mechanism by which C/EBPα regulates PPARγ expression by acetylating histones and revealed the epigenetic phenomenon in SANFH for the first time.
Collapse
Affiliation(s)
- Ping Duan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hanyu Wang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xinzeyu Yi
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hao Zhang
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Hui Chen
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhenyu Pan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
29
|
Rashid S, Anjum S, Ahmad A, Nadeem R, Ahmed M, Shah SAA, Abdullah M, Zia K, Ul-haq Z. Betamethasone Dipropionate Derivatization, Biotransformation, Molecular Docking, and ADME Analysis as Glucocorticoid Receptor. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6865472. [PMID: 35865666 PMCID: PMC9296322 DOI: 10.1155/2022/6865472] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/14/2022] [Indexed: 11/26/2022]
Abstract
Betamethasone is an important glucocorticoids (GCs), frequently used to cure allergies (such as asthma and angioedema), Crohn's disease, skin diseases (such as dermatitis and psoriasis), systemic lupus erythematosus, rheumatic disorders, and leukemia. Present investigation deals to find potential agonist of glucocorticoid receptors after biotransformation of betamethasone dipropionate (1) and to carry out the molecular docking and ADME analyses. Biotransformation of 1 was carried out with Launaea capitata (dandy) roots and Musa acuminate (banana) leaves. M. acuminate furnished low-cost value-added products such as Sananone dipropionate (2) in 5% yields. Further, biocatalysis of Sananone dipropionate (2) with M. acuminate gave Sananone propionate (3) and Sananone (4) in 12% and 7% yields, respectively. However, Sananone (4) was obtained in 37% yields from Launaea capitata. Compound 5 was obtained in 11% yield after β-elimination of propionic acid at C-17 during oxidation of compound 1. The structure elucidation of new compounds 2-5 was accomplished through combined use of X-ray diffraction and NMR (1D and 2D) studies. In addition to this, molecular docking and ADME analyses of all transformed products of 1 were also done. Compounds 1-5 showed -12.53 to -10.11 kcal/mol potential binding affinity with glucocorticoid receptor (GR) and good ADME profile. Moreover, all the compounds showed good oral bioavailability with the octanol/water partition coefficient in the range of 2.23 to 3.65, which indicated that compounds 1-5 were in significant agreement with the given criteria to be considered as drug-like.
Collapse
Affiliation(s)
- Sana Rashid
- Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Shazia Anjum
- Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
- H.E.J. Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Aqeel Ahmad
- University of Chinese Academy of Science (UCAS), Beijing, China
| | - Raziya Nadeem
- Department of Chemistry, University of Agriculture, Faisalabad, Pakistan
| | - Maqsood Ahmed
- Material Chemistry Lab, Institute of Chemistry, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Syed Adnan Ali Shah
- Faculty of Pharmacy, Atta-ur-Rahman Institute for Natural Products Discovery, Universiti Teknologi MARA, Cawangan Selangor Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor D. E, Malaysia
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Universiti Teknologi MARA Cawangan Selangor Kampus Puncak Alam, 2300 Bandar Puncak Alam, Selangor, Malaysia
| | - Muhammad Abdullah
- Cholistan Institute of Desert Studies, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Komal Zia
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Zaheer Ul-haq
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
30
|
Zou H, Ye H, Zhang J, Ren L. Recent advances in nuclear receptors-mediated health benefits of blueberry. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 100:154063. [PMID: 35344717 DOI: 10.1016/j.phymed.2022.154063] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Blueberry is rich in bioactive substances and has anti-oxidant, anti-inflammatory, anti-obesity, anti-cancer, neuroprotective, and other activities. Blueberry has been shown to treat diseases by mediating the transcription of nuclear receptors. However, evidence for nuclear receptor-mediated health benefits of blueberry has not been systematically reviewed. PURPOSE This review aims to summarize the nuclear receptor-mediated health benefits of blueberry. METHODS This study reviews all relevant literature published in NCBI PubMed, Scopus, Web of Science, and Google Scholar by January 2022. The relevant literature was extracted from the databases with the following keyword combinations: "biological activities" OR "nuclear receptors" OR "phytochemicals" AND "blueberry" OR "Vaccinium corymbosum" as well as free-text words. RESULTS In vivo and in vitro experimental results and clinical evidence have demonstrated that blueberry has health-promoting effects. Supplementing blueberry is beneficial to the treatment of cancer, the alleviation of metabolic syndrome, and liver protection. Blueberry can regulate the transcription of PPARs, ERs, AR, GR, MR, LXRs, and FXR and mediate the expressions of Akt, CYP 1Al, p53, and Bcl-2. CONCLUSION Blueberry can be targeted to treat various diseases by mediating the transcription of nuclear receptors. Nevertheless, further human research is needed.
Collapse
Affiliation(s)
- Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Haiqing Ye
- College of Food Science and Engineering, Jilin University, Changchun 130062, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| | - Li Ren
- College of Food Science and Engineering, Jilin University, Changchun 130062, China.
| |
Collapse
|
31
|
Kośmider K, Karska K, Kozakiewicz A, Lejman M, Zawitkowska J. Overcoming Steroid Resistance in Pediatric Acute Lymphoblastic Leukemia-The State-of-the-Art Knowledge and Future Prospects. Int J Mol Sci 2022; 23:ijms23073795. [PMID: 35409154 PMCID: PMC8999045 DOI: 10.3390/ijms23073795] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 12/13/2022] Open
Abstract
Acute lymphoblastic leukemia (ALL) is the most common malignancy among children. Despite the enormous progress in ALL therapy, resulting in achieving a 5-year survival rate of up to 90%, the ambitious goal of reaching a 100% survival rate is still being pursued. A typical ALL treatment includes three phases: remission induction and consolidation and maintenance, preceded by a prednisone prephase. Poor prednisone response (PPR) is defined as the presence of ≥1.0 × 109 blasts/L in the peripheral blood on day eight of therapy and results in significantly frequent relapses and worse outcomes. Hence, identifying risk factors of steroid resistance and finding methods of overcoming that resistance may significantly improve patients' outcomes. A mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK-ERK) pathway seems to be a particularly attractive target, as its activation leads to steroid resistance via a phosphorylating Bcl-2-interacting mediator of cell death (BIM), which is crucial in the steroid-induced cell death. Several mutations causing activation of MAPK-ERK were discovered, notably the interleukin-7 receptor (IL-7R) pathway mutations in T-cell ALL and rat sarcoma virus (Ras) pathway mutations in precursor B-cell ALL. MAPK-ERK pathway inhibitors were demonstrated to enhance the results of dexamethasone therapy in preclinical ALL studies. This report summarizes steroids' mechanism of action, resistance to treatment, and prospects of steroids therapy in pediatric ALL.
Collapse
Affiliation(s)
- Kamil Kośmider
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland; (K.K.); (A.K.)
| | - Katarzyna Karska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland;
| | - Agata Kozakiewicz
- Student Scientific Society, Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland; (K.K.); (A.K.)
| | - Monika Lejman
- Laboratory of Genetic Diagnostics, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Gębali 6, 20-093 Lublin, Poland;
- Correspondence:
| |
Collapse
|
32
|
Wang H, Kan WJ, Feng Y, Feng L, Yang Y, Chen P, Xu JJ, Si TM, Zhang L, Wang G, Du J. Nuclear receptors modulate inflammasomes in the pathophysiology and treatment of major depressive disorder. World J Psychiatry 2021; 11:1191-1205. [PMID: 35070770 PMCID: PMC8717028 DOI: 10.5498/wjp.v11.i12.1191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 07/29/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Major depressive disorder (MDD) is highly prevalent and is a significant cause of mortality and morbidity worldwide. Currently, conventional pharmacological treatments for MDD produce temporary remission in < 50% of patients; therefore, there is an urgent need for a wider spectrum of novel antidepressants to target newly discovered underlying disease mechanisms. Accumulated evidence has shown that immune inflammation, particularly inflammasome activity, plays an important role in the pathophysiology of MDD. In this review, we summarize the evidence on nuclear receptors (NRs), such as glucocorticoid receptor, mineralocorticoid receptor, estrogen receptor, aryl hydrocarbon receptor, and peroxisome proliferator-activated receptor, in modulating the inflammasome activity and depression-associated behaviors. This review provides evidence from an endocrine perspective to understand the role of activated NRs in the pathophysiology of MDD, and to provide insight for the discovery of antidepressants with novel mechanisms for this devastating disorder.
Collapse
Affiliation(s)
- Han Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Wei-Jing Kan
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Yuan Feng
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Lei Feng
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Yang Yang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Pei Chen
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Jing-Jie Xu
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Tian-Mei Si
- Department of Clinical Psychopharmacology, Peking University Institute of Mental Health, Beijing 100191, Beijing Province, China
| | - Ling Zhang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Gang Wang
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
| | - Jing Du
- The National Clinical Research Center for Mental Disorders & Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Beijing 100088, Beijing Province, China
- State Key Laboratory for Conservation and Utilization of Bio-resources in Yunnan, Yunnan University, Kunming 650091, Yunnan Province, China
| |
Collapse
|
33
|
Xu S, Liu Y, Hu R, Wang M, Stöhr O, Xiong Y, Chen L, Kang H, Zheng L, Cai S, He L, Wang C, Copps KD, White MF, Miao J. TAZ inhibits glucocorticoid receptor and coordinates hepatic glucose homeostasis in normal physiological states. eLife 2021; 10:e57462. [PMID: 34622775 PMCID: PMC8555985 DOI: 10.7554/elife.57462] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/13/2021] [Indexed: 12/11/2022] Open
Abstract
The elucidation of the mechanisms whereby the liver maintains glucose homeostasis is crucial for the understanding of physiological and pathological states. Here, we show a novel role of hepatic transcriptional co-activator with PDZ-binding motif (TAZ) in the inhibition of glucocorticoid receptor (GR). TAZ is abundantly expressed in pericentral hepatocytes and its expression is markedly reduced by fasting. TAZ interacts via its WW domain with the ligand-binding domain of GR to limit the binding of GR to the GR response element in gluconeogenic gene promoters. Therefore, liver-specific TAZ knockout mice show increases in glucose production and blood glucose concentration. Conversely, the overexpression of TAZ in mouse liver reduces the binding of GR to gluconeogenic gene promoters and glucose production. Thus, our findings demonstrate that hepatic TAZ inhibits GR transactivation of gluconeogenic genes and coordinates gluconeogenesis in response to physiological fasting and feeding.
Collapse
Affiliation(s)
- Simiao Xu
- Division of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Branch of the National Clinical Research Center for Metabolic DiseaseWuhanChina
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Yangyang Liu
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Ruixiang Hu
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Min Wang
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Department of Pathology, Beth Israel Deaconess Medical CenterBostonUnited States
| | - Oliver Stöhr
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Yibo Xiong
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Liang Chen
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- College of Science, Northeastern UniversityBostonUnited States
| | - Hong Kang
- Department of Systemic Biology, Harvard Medical SchoolBostonUnited States
| | - Lingyun Zheng
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Songjie Cai
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Transplantation Research Center, Brigham and Women’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Li He
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Cunchuan Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Kyle D Copps
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Morris F White
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Ji Miao
- Division of Endocrinology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
- Department of Pediatrics, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
34
|
Mahadik N, Bhattacharya D, Padmanabhan A, Sakhare K, Narayan KP, Banerjee R. Targeting steroid hormone receptors for anti-cancer therapy-A review on small molecules and nanotherapeutic approaches. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 14:e1755. [PMID: 34541822 DOI: 10.1002/wnan.1755] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
The steroid hormone receptors (SHRs) among nuclear hormone receptors (NHRs) are steroid ligand-dependent transcription factors that play important roles in the regulation of transcription of genes promoted via hormone responsive elements in our genome. Aberrant expression patterns and context-specific regulation of these receptors in cancer, have been routinely reported by multiple research groups. These gave an window of opportunity to target those receptors in the context of developing novel, targeted anticancer therapeutics. Besides the development of a plethora of SHR-targeting synthetic ligands and the availability of their natural, hormonal ligands, development of many SHR-targeted, anticancer nano-delivery systems and theranostics, especially based on small molecules, have been reported. It is intriguing to realize that these cytoplasmic receptors have become a hot target for cancer selective delivery. This is in spite of the fact that these receptors do not fall in the category of conventional, targetable cell surface bound or transmembrane receptors that enjoy over-expression status. Glucocorticoid receptor (GR) is one such exciting SHR that in spite of it being expressed ubiquitously in all cells, we discovered it to behave differently in cancer cells, thus making it a truly druggable target for treating cancer. This review selectively accumulates the knowledge generated in the field of SHR-targeting as a major focus for cancer treatment with various anticancer small molecules and nanotherapeutics on progesterone receptor, mineralocorticoid receptor, and androgen receptor while selectively emphasizing on GR and estrogen receptor. This review also briefly highlights lipid-modification strategy to convert ligands into SHR-targeted cancer nanotherapeutics. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Namita Mahadik
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Dwaipayan Bhattacharya
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Akshaya Padmanabhan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kalyani Sakhare
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Kumar Pranav Narayan
- Department of Biological Sciences, Birla Institute of Technology Pilani, Hyderabad, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad, India.,Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
35
|
Myung CH, Lee JE, Jo CS, Park JI, Hwang JS. Regulation of Melanophilin (Mlph) gene expression by the glucocorticoid receptor (GR). Sci Rep 2021; 11:16813. [PMID: 34413386 PMCID: PMC8376885 DOI: 10.1038/s41598-021-96276-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 08/06/2021] [Indexed: 02/07/2023] Open
Abstract
Mlph plays a crucial role in regulating skin pigmentation through the melanosome transport process. Although Mlph is a major component involved in melanosome transport, the mechanism that regulates the expression of the Mlph gene has not been identified. In this study, we demonstrate that Mlph expression is regulated by the glucocorticoid receptor (GR). Alteration of GR activity using a specific GR agonist or antagonist only regulated the expression of Mlph among the 3 key melanosome transport proteins. Translocation of GR from the cytosol into the nucleus following Dex treatment was confirmed by separating the cytosol and nuclear fractions and by immunofluorescence staining. In ChIP assays, Dex induced GR binding to the Mlph promoter and we determined that Dex induced the GR binding motif on the Mlph promoter. Our findings contribute to understanding the regulation of Mlph expression and to the novel role of GR in Mlph gene expression.
Collapse
Affiliation(s)
- Cheol Hwan Myung
- Department of Genetic Engineering & Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Gyeonggi-do, 17104, Republic of Korea
| | - Ji Eun Lee
- Department of Genetic Engineering & Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Gyeonggi-do, 17104, Republic of Korea
| | - Chan Song Jo
- Department of Genetic Engineering & Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Gyeonggi-do, 17104, Republic of Korea
| | - Jong Il Park
- Department of Genetic Engineering & Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Gyeonggi-do, 17104, Republic of Korea
| | - Jae Sung Hwang
- Department of Genetic Engineering & Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Gyeonggi-do, 17104, Republic of Korea.
| |
Collapse
|
36
|
Abstract
Background: Acute lymphoblastic leukemia (ALL) is a malignant disease characterized by an excessive number of immature lymphocytes, including immature precursors of both B- and T cells. ALL affects children more often than adults. Immature lymphocytes lead to arrested differentiation and proliferation of cells. Its conventional treatments involve medication with dexamethasone, vincristine, and other anticancer drugs. Although the current first-line drugs can achieve effective treatment, they still cannot prevent the recurrence of some patients with ALL. Treatments have high risk of recurrence especially after the first remission. Currently, novel therapies to treat ALL are in need. Autophagy and apoptosis play important roles in regulating cancer development. Autophagy involves degradation of proteins and organelles, and apoptosis leads to cell death. These phenomena are crucial in cancer progression. Past studies reported that many potential anticancer agents regulate intracellular signaling pathways. Methods: The authors discuss the recent research findings on the role of autophagy and apoptosis in ALL. Results: The autophagy and apoptosis are widely used in the treatment of ALL. Most studies showed that many agents regulate autophagy and apoptosis in ALL cell models, clinical trials, and ALL animal models. Conclusions: In summary, activating autophagy and apoptosis pathways are the main strategies for ALL treatments. For ALL, combining new drugs with traditional chemotherapy and glucocorticoids treatments can achieve the greatest therapeutic effect by activating autophagy and apoptosis.
Collapse
Affiliation(s)
- Fang-Liang Huang
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC.,Department of Physical Therapy, Hungkuang University, Taichung, Taiwan, ROC.,Institute of Biomedical Sciences, MacKay Medical College, New Taipei City, Taiwan, ROC
| | - Sheng-Jie Yu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan, ROC
| | - Chia-Ling Li
- Children's Medical Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| |
Collapse
|
37
|
Manjur ABMK, Lempiäinen JK, Malinen M, Varjosalo M, Palvimo JJ, Niskanen EA. BCOR modulates transcriptional activity of a subset of glucocorticoid receptor target genes involved in cell growth and mobility. J Steroid Biochem Mol Biol 2021; 210:105873. [PMID: 33722704 DOI: 10.1016/j.jsbmb.2021.105873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/29/2022]
Abstract
Glucocorticoid (GC) receptor (GR) is a key transcription factor (TF) that regulates vital metabolic and anti-inflammatory processes. We have identified BCL6 corepressor (BCOR) as a dexamethasone-stimulated interaction partner of GR. BCOR is a component of non-canonical polycomb repressor complex 1.1 (ncPCR1.1) and linked to different developmental disorders and cancers, but the role of BCOR in GC signaling is poorly characterized. Here, using ChIP-seq we show that, GC induces genome-wide redistribution of BCOR chromatin binding towards GR-occupied enhancers in HEK293 cells. As assessed by RNA-seq, depletion of BCOR altered the expression of hundreds of GC-regulated genes, especially the ones linked to TNF signaling, GR signaling and cell migration pathways. Biotinylation-based proximity mapping revealed that GR and BCOR share several interacting partners, including nuclear receptor corepressor NCOR1. ChIP-seq showed that the NCOR1 co-occurs with both BCOR and GR on a subset of enhancers upon GC treatment. Simultaneous depletion of BCOR and NCOR1 influenced GR target gene expression in a combinatorial and gene-specific manner. Finally, we show using live cell imaging that the depletion of BCOR together with NCOR1 markedly enhances cell migration. Collectively, our data suggest BCOR as an important gene and pathway selective coregulator of GR transcriptional activity.
Collapse
Affiliation(s)
| | | | - Marjo Malinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland; Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Markku Varjosalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland
| | - Einari A Niskanen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
38
|
Forecasting early onset diminished ovarian reserve for young reproductive age women. J Assist Reprod Genet 2021; 38:1853-1860. [PMID: 33786734 DOI: 10.1007/s10815-021-02155-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/14/2021] [Indexed: 10/21/2022] Open
Abstract
PURPOSE To investigate the biological networks associated with DOR in young women and the subsequent molecular impact on preimplantation embryos. METHODS Whole peripheral blood was collected from patients: young women presenting with diminished ovarian reserve (DOR) and age-matched young women with normal ovarian reserve. Maternal exome sequencing was performed on the NovaSEQ 6000 and sequencing validation was completed using Taqman® SNP Genotyping Assays. Blastocyst global methylome and transcriptome sequencing were also analyzed. RESULTS Exome sequencing revealed 730 significant DNA variants observed exclusively in the young DOR patients. Bioinformatic analysis revealed a significant impact to the Glucocorticoid receptor (GR) signaling pathway and each young DOR female had an average of 6.2 deleterious DNA variants within this pathway. Additional stratification based on patient age resulted in a cut-off at 31 years for young DOR discrimination. Embryonic global methylome sequencing resulted in only a very small number of total CpG sites with methylation alterations (1,775; 0.015% of total) in the DOR group. Additionally, there was no co-localization between these limited number of altered CpG sites and significant variants, genes, or pathways. RNA sequencing also resulted in no biologically significant transcription changes between DOR blastocysts and controls. CONCLUSION GR signaling DNA variants were observed in women with early-onset DOR potentially compromising oocyte production and quality. However, no significant downstream effects on biological processes appear to impact the resulting blastocyst. The ability to forecast premature DOR for young women may allow for earlier identification and clinical intervention for this patient population.
Collapse
|
39
|
Gromotowicz-Poplawska A, Szoka P, Zakrzeska A, Kolodziejczyk P, Marcinczyk N, Szemraj J, Tutka P, Chabielska E. Hyperglycemia Potentiates Prothrombotic Effect of Aldosterone in a Rat Arterial Thrombosis Model. Cells 2021; 10:cells10020471. [PMID: 33671798 PMCID: PMC7927020 DOI: 10.3390/cells10020471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 11/16/2022] Open
Abstract
We investigated the role of aldosterone (ALDO) in the development of arterial thrombosis in streptozotocin-induced diabetic rats. To evaluate the effect of endogenous ALDO, the rats underwent adrenalectomy (ADX). ADX reduced the development of arterial thrombosis. A 1 h infusion of ALDO (30 μg/kg/h) enhanced thrombosis in adrenalectomized rats, while this effect was potentiated in diabetic rats. ALDO shortened bleeding time, increased plasma levels of tissue factor (TF) and plasminogen activator inhibitor, decreased plasma level of nitric oxide (NO) metabolites, and increased oxidative stress. Moreover, 2 h incubation of human umbilical vein endothelial cells (HUVECs) with ALDO (10-7 M) disrupted hemostatic balance in endothelial cells in normoglycemia (glucose 5.5 mM), and this effect was more pronounced in hyperglycemia (glucose 30 mM). We demonstrated that the acute ALDO infusion enhances arterial thrombosis in rats and hyperglycemia potentiates this prothrombotic effect. The mechanism of ALDO action was partially mediated by mineralocorticoid (MR) and glucocorticoid (GR) receptors and related to impact of the hormone on primary hemostasis, TF-dependent coagulation cascade, fibrinolysis, NO bioavailability, and oxidative stress balance. Our in vitro study confirmed that ALDO induces prothrombotic phenotype in the endothelium, particularly under hyperglycemic conditions.
Collapse
Affiliation(s)
- Anna Gromotowicz-Poplawska
- Department of Biopharmacy, Medical University of Bialystok, 15-222 Bialystok, Poland; (N.M.); (E.C.)
- Correspondence: ; Tel.: +48-857485804
| | - Piotr Szoka
- Department of Pharmacology, Medical University of Bialystok, 15-222 Bialystok, Poland;
| | | | - Patrycjusz Kolodziejczyk
- Department of Experimental and Clinical Pharmacology, University of Rzeszow, 35-959 Rzeszow, Poland; (P.K.); (P.T.)
| | - Natalia Marcinczyk
- Department of Biopharmacy, Medical University of Bialystok, 15-222 Bialystok, Poland; (N.M.); (E.C.)
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Piotr Tutka
- Department of Experimental and Clinical Pharmacology, University of Rzeszow, 35-959 Rzeszow, Poland; (P.K.); (P.T.)
- National Drug and Alcohol Research Center, University of New South Wales, Sydney 2052, Australia
| | - Ewa Chabielska
- Department of Biopharmacy, Medical University of Bialystok, 15-222 Bialystok, Poland; (N.M.); (E.C.)
| |
Collapse
|
40
|
Zarantoniello M, Bortoletti M, Olivotto I, Ratti S, Poltronieri C, Negrato E, Caberlotto S, Radaelli G, Bertotto D. Salinity, Temperature and Ammonia Acute Stress Response in Seabream ( Sparus aurata) Juveniles: A Multidisciplinary Study. Animals (Basel) 2021; 11:E97. [PMID: 33419050 PMCID: PMC7825456 DOI: 10.3390/ani11010097] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/30/2020] [Accepted: 01/02/2021] [Indexed: 11/16/2022] Open
Abstract
The present study aimed to investigate the acute response of gilthead seabream (Sparus aurata) juveniles exposed to temperature, salinity and ammonia stress. Radioimmunoassay was used to evaluate cortisol levels, whereas insulin-like growth factors (igf1 and igf2), myostatin (mstn), heat-shock protein 70 (hsp70) and glucocorticoid receptor (gr) gene expression was assessed trough Real-Time PCR. The presence and localization of IGF-I and HSP70 were investigated by immunohistochemistry. In all the stress conditions, a significant increase in cortisol levels was observed reaching higher values in the thermic and chemical stress groups. Regarding fish growth markers, igf1 gene expression was significantly higher only in fish subjected to heat shock stress while, at 60 min, igf2 gene expression was significantly lower in all the stressed groups. Temperature and ammonia changes resulted in a higher mstn gene expression. Molecular analyses on stress response evidenced a time dependent increase in hsp70 gene expression, that was significantly higher at 60 min in fish exposed to heat shock and chemical stress. Furthermore, the same experimental groups were characterized by a significantly higher gr gene expression respect to the control one. Immunostaining for IGF-I and HSP70 antibodies was observed in skin, gills, liver, and digestive system of gilthead seabream juveniles.
Collapse
Affiliation(s)
- Matteo Zarantoniello
- Department of Life and Environmental Sciences, Polytechnic University of Marche, I-60131 Ancona, Italy; (M.Z.); (I.O.); (S.R.)
| | - Martina Bortoletti
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Viale dell’Università 16, I-35020 Legnaro, Padova, Italy; (M.B.); (C.P.); (E.N.); (D.B.)
| | - Ike Olivotto
- Department of Life and Environmental Sciences, Polytechnic University of Marche, I-60131 Ancona, Italy; (M.Z.); (I.O.); (S.R.)
| | - Stefano Ratti
- Department of Life and Environmental Sciences, Polytechnic University of Marche, I-60131 Ancona, Italy; (M.Z.); (I.O.); (S.R.)
| | - Carlo Poltronieri
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Viale dell’Università 16, I-35020 Legnaro, Padova, Italy; (M.B.); (C.P.); (E.N.); (D.B.)
| | - Elena Negrato
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Viale dell’Università 16, I-35020 Legnaro, Padova, Italy; (M.B.); (C.P.); (E.N.); (D.B.)
| | - Stefano Caberlotto
- Valle Ca’ Zuliani Società Agricola Srl, I-34074 Monfalcone, Gorizia, Italy;
| | - Giuseppe Radaelli
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Viale dell’Università 16, I-35020 Legnaro, Padova, Italy; (M.B.); (C.P.); (E.N.); (D.B.)
| | - Daniela Bertotto
- Department of Comparative Biomedicine and Food Science (BCA), University of Padova, Viale dell’Università 16, I-35020 Legnaro, Padova, Italy; (M.B.); (C.P.); (E.N.); (D.B.)
| |
Collapse
|
41
|
Sridharan K, Rathore B, Yousuf M, Reddy Rachamalla HK, Jinka S, Jaggarapu MMCS, Banerjee R. Self-Assembling Derivative of Hydrocortisone as Glucocorticoid Receptor-Targeted Nanotherapeutics for Synergistic, Combination Therapy against Colorectal Tumor. Mol Pharm 2020; 18:1208-1228. [PMID: 33371687 DOI: 10.1021/acs.molpharmaceut.0c01091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Hydrocortisone, a natural glucocorticoid secreted by adrenal and extra-adrenal tissues, locally governs the transcription of genes involved in inflammation, immune response, metabolism, and energy homeostasis via binding to its cognate glucocorticoid receptor (GR). In this study, we show that modified hydrocortisone (HC16), a cancer-selective cytotoxic molecule, showed synergism in combination with drugs like Doxorubicin and docetaxel, self-assembled into vesicles, entrapped docetaxel and complexed with anti-cancer plasmid DNA for enhanced killing of cancer cells. These vesicles exhibited GR-mediated nuclear localization, delivery of the p53 gene, and also inhibited cell viability selectively in RKO, HCT15, and CT26 colon cancer cells but not in normal cells like CHO and HEK293T. Apart from exerting its own anti-cancer activity, the self-assembled HC16 vesicles loaded with docetaxel sensitized the cancer cells to its drug cargo by downregulating the drug metabolizing CYP3A4 gene. This indirectly reduces the risk of nonspecific adverse effects in normal cells, as the viability of sensitized cancer cells could be significantly reduced even in low doses of cytotoxic docetaxel. The near infrared (NIR)-dye-associated self-assemblies accumulated in a colon tumor with higher orders of NIR intensity compared to those in a colon of healthy mice. Thereafter, the treatment of HC16-docetaxel-p53 vesicle/DNA complex led to significant tumor regression, which resulted in a cecum/body weight ratio in tumor-bearing mice similar to that of healthy mice measured at 24 h postcompletion of treatment. There was an up to 2.5-fold enhancement in the overall survivability of colon-tumor-bearing mice treated with HC16-docetaxel-p53 vesicle/DNA complexes when compared against the pristine docetaxel-treated groups. Further, the HC16-docetaxel-p53 vesicle/DNA complex-treated group showed reduced nuclear accumulation of cell proliferation marker Ki67, reduced protein levels of prosurvival and mesenchymal proteins like Bcl-2, PARP, vimentin, and N-cadherin, and increased the levels of pro-apoptotic activated caspases as compared to the pristine docetaxel-treated groups. The therapeutic package described herein is expected to find future use as a rational, multifaceted, GR-targeted approach for inhibiting colon tumor progression.
Collapse
Affiliation(s)
- Kathyayani Sridharan
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Bhowmira Rathore
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India
| | - Md Yousuf
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Department of Chemistry, Ramnagar College, Purba Medinipur, West Bengal 721 453, India
| | - Hari Krishna Reddy Rachamalla
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Sudhakar Jinka
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Madhan Mohan Chandra Sekhar Jaggarapu
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| | - Rajkumar Banerjee
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 007, India.,Academy of Scientific and Innovative Research (AcSIR), CSIR-HRDC Campus, Ghaziabad 201002, India
| |
Collapse
|
42
|
Haleem DJ. Glucocorticoids in the Physiological and Transcriptional Regulation of 5-HT1A Receptor and the Pathogenesis of Depression. Neuroscientist 2020; 28:59-68. [PMID: 33243080 DOI: 10.1177/1073858420975711] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is growing increase in the global prevalence of depression, but treatment outcome of this highly disabling disease is not satisfactory. Many patients are not benefitted by the currently prescribed antidepressants-together with this partial remission is very common. Improving treatment strategies and developing better therapeutic agents for treating depression is therefore highly needed. Stress-related epigenetic changes play a critical role in the pathogenesis as well as treatment of depression. Stressful events activate hypothalamic-pituitary-adrenal axis to increase circulating levels of glucocorticoids (GCs), and a greater sensitivity to this fright and flight response increases risk of depression. A role of serotonin (5-hydroxytryptamine; 5-HT) in responses to stress and in the pathogenesis and treatment of depression is well established. Substantial evidence supports a critical role of 5-HT1A receptors in these effects of 5-HT. The present article targets stress-induced higher and sustained increases of GCs and mediated influences on the physiological as well transcriptional regulation of 5-HT1A receptors to evaluate their causal role in the pathogenesis of depression. It is suggested that synthetic compounds with antagonist activity for GC receptors and agonist activity for 5-HT1A receptors may prove better therapeutic agents for treating depression.
Collapse
Affiliation(s)
- Darakhshan Jabeen Haleem
- Neuroscience Research Laboratory, Dr Panjwani Center for Molecular Medicine & Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi, Pakistan
| |
Collapse
|
43
|
Bae S, Zeng S, Park-Min KH. Nuclear receptors in osteoclasts. Curr Opin Pharmacol 2020; 53:8-17. [PMID: 32569976 PMCID: PMC7669703 DOI: 10.1016/j.coph.2020.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 02/08/2023]
Abstract
Osteoclasts are bone-resorbing cells that play an essential role in the remodeling of bone under physiological conditions and numerous pathological conditions, such as osteoporosis, bone metastasis, and inflammatory bone erosion. Nuclear receptors are crucial to various physiological processes, including metabolism, development and inflammation, and function as transcription factors to activate target genes. Synthetic ligands of nuclear receptors are also available for the treatment of metabolic and inflammatory diseases. However, dysregulated bone phenotypes have been documented in patients who take synthetic nuclear receptor ligands as a therapy. Therefore, the effect of nuclear receptors on bone cells has become an important area of exploration; additionally, the molecular mechanisms underlying the action of nuclear receptors in osteoclasts have not been completely understood. Here, we cover the recent progress in our understanding of the roles of nuclear receptors in osteoclasts.
Collapse
Affiliation(s)
- Seyeon Bae
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Steven Zeng
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA
| | - Kyung-Hyun Park-Min
- Arthritis and Tissue Degeneration Program, David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY 10021, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA; BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10021, USA.
| |
Collapse
|
44
|
Adrenal insufficiency: Physiology, clinical presentation and diagnostic challenges. Clin Chim Acta 2020; 505:78-91. [PMID: 32035851 DOI: 10.1016/j.cca.2020.01.029] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/24/2020] [Accepted: 01/27/2020] [Indexed: 12/21/2022]
Abstract
Adrenal insufficiency (AI) is a serious condition, which can arise from pathology affecting the adrenal gland itself (primary adrenal insufficiency, PAI), hypothalamic or pituitary pathology (secondary adrenal insufficiency, SAI), or as a result of suppression of the hypothalamic-pituitaryadrenal (HPA) axis by exogenous glucocorticoid therapy (tertiary adrenal insufficiency, TAI). AI is associated with an increase in morbidity and mortality and a reduction in quality of life. In addition, the most common cause of PAI, autoimmune adrenalitis, may be associated with a variety of other autoimmune disorders. Untreated AI can present with chronic fatigue, weight loss and vulnerability to infection. The inability to cope with acute illness or infection can precipitate life-threatening adrenal crisis. It is therefore a critical diagnosis to make in a timely fashion, in order to institute appropriate management, aimed at reversing chronic ill health, preventing acute crises, and restoring quality of life. In this review, we will describe the normal physiology of the HPA axis and explain how knowledge of the physiology of this axis helps us understand the clinical presentation of AI, and forms the basis for the biochemical investigations which lead to the diagnosis of AI.
Collapse
|
45
|
Hox V, Lourijsen E, Jordens A, Aasbjerg K, Agache I, Alobid I, Bachert C, Boussery K, Campo P, Fokkens W, Hellings P, Hopkins C, Klimek L, Mäkelä M, Mösges R, Mullol J, Pujols L, Rondon C, Rudenko M, Toppila-Salmi S, Scadding G, Scheire S, Tomazic PV, Van Zele T, Wagemann M, van Boven JFM, Gevaert P. Benefits and harm of systemic steroids for short- and long-term use in rhinitis and rhinosinusitis: an EAACI position paper. Clin Transl Allergy 2020; 10:1. [PMID: 31908763 PMCID: PMC6941282 DOI: 10.1186/s13601-019-0303-6] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 12/02/2019] [Indexed: 02/05/2023] Open
Abstract
Because of the inflammatory mechanisms of most chronic upper airway diseases such as rhinitis and chronic rhinosinusitis, systemic steroids have been used for their treatment for decades. However, it has been very well documented that—potentially severe—side-effects can occur with the accumulation of systemic steroid courses over the years. A consensus document summarizing the benefits of systemic steroids for each upper airway disease type, as well as highlighting the potential harms of this treatment is currently lacking. Therefore, a panel of international experts in the field of Rhinology reviewed the available literature with the aim of providing recommendations for the use of systemic steroids in treating upper airway disease.
Collapse
Affiliation(s)
- Valerie Hox
- 1Cliniques Universitaires Saint-Luc Brussels, Av. Hippocrate 10, 1200 Brussels, Belgium
| | - Evelijn Lourijsen
- 2Department of Otorhinolaryngology, Amsterdam University Medical Centres, AMC, Amsterdam, The Netherlands
| | - Arnout Jordens
- 3Upper Airway Research Laboratory, Dep. of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | | | - Ioana Agache
- Faculty of Medicine, Transsylvania University, Brasov, Romania
| | - Isam Alobid
- 6Hospital Clínic, IDIBAPS, CEBERES Universitat de Barcelona, Catalonia, Spain.,7Centro Medico Teknon, Barcelona, Spain
| | - Claus Bachert
- 3Upper Airway Research Laboratory, Dep. of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium.,8Department of Ear, Nose and Throat Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Koen Boussery
- 9Pharmaceutical Care Unit, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Paloma Campo
- 10Allergy Unit, Hospital Regional Universitario of Málaga, IBIMA, ARADyAL, Malaga, Spain
| | - Wytske Fokkens
- 2Department of Otorhinolaryngology, Amsterdam University Medical Centres, AMC, Amsterdam, The Netherlands
| | - Peter Hellings
- 11Department of Ear, Nose and Throat Disease, University Hospitals, Louvain, Belgium
| | - Claire Hopkins
- 12ENT Department, Guy's & St Thomas' Hospital, London, UK
| | - Ludger Klimek
- Center of Rhinology and Allergology, Wiesbaden, Germany
| | - Mika Mäkelä
- 14Skin and Allergy Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | - Joaquim Mullol
- 6Hospital Clínic, IDIBAPS, CEBERES Universitat de Barcelona, Catalonia, Spain
| | - Laura Pujols
- 6Hospital Clínic, IDIBAPS, CEBERES Universitat de Barcelona, Catalonia, Spain
| | - Carmen Rondon
- 10Allergy Unit, Hospital Regional Universitario of Málaga, IBIMA, ARADyAL, Malaga, Spain
| | | | - Sanna Toppila-Salmi
- 14Skin and Allergy Hospital, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | | | - Sophie Scheire
- 9Pharmaceutical Care Unit, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | | | - Thibaut Van Zele
- 3Upper Airway Research Laboratory, Dep. of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | | | - Job F M van Boven
- 20Department of Clinical Pharmacy & Pharmacology, University Medical Center Groningen, Groningen Research Institute for Asthma and COPD (GRIAC), University of Groningen, Groningen, The Netherlands
| | - Philippe Gevaert
- 3Upper Airway Research Laboratory, Dep. of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
46
|
Ahmad M, Hachemi Y, Paxian K, Mengele F, Koenen M, Tuckermann J. A Jack of All Trades: Impact of Glucocorticoids on Cellular Cross-Talk in Osteoimmunology. Front Immunol 2019; 10:2460. [PMID: 31681333 PMCID: PMC6811614 DOI: 10.3389/fimmu.2019.02460] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 10/02/2019] [Indexed: 12/13/2022] Open
Abstract
Glucocorticoids (GCs) are known to have a strong impact on the immune system, metabolism, and bone homeostasis. While these functions have been long investigated separately in immunology, metabolism, or bone biology, the understanding of how GCs regulate the cellular cross-talk between innate immune cells, mesenchymal cells, and other stromal cells has been garnering attention rather recently. Here we review the recent findings of GC action in osteoporosis, inflammatory bone diseases (rheumatoid and osteoarthritis), and bone regeneration during fracture healing. We focus on studies of pre-clinical animal models that enable dissecting the role of GC actions in innate immune cells, stromal cells, and bone cells using conditional and function-selective mutant mice of the GC receptor (GR), or mice with impaired GC signaling. Importantly, GCs do not only directly affect cellular functions, but also influence the cross-talk between mesenchymal and immune cells, contributing to both beneficial and adverse effects of GCs. Given the importance of endogenous GCs as stress hormones and the wide prescription of pharmaceutical GCs, an improved understanding of GC action is decisive for tackling inflammatory bone diseases, osteoporosis, and aging.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Yasmine Hachemi
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Kevin Paxian
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Florian Mengele
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
- Praxisklinik für Orthopädie, Unfall- und Neurochirurgie Prof. Bischoff/ Dr. Spies/ Dr. Mengele, Neu-Ulm, Germany
| | - Mascha Koenen
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, United States
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| |
Collapse
|
47
|
Spulber S, Conti M, Elberling F, Raciti M, Borroto-Escuela DO, Fuxe K, Ceccatelli S. Desipramine restores the alterations in circadian entrainment induced by prenatal exposure to glucocorticoids. Transl Psychiatry 2019; 9:263. [PMID: 31624238 PMCID: PMC6797805 DOI: 10.1038/s41398-019-0594-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/09/2019] [Accepted: 09/24/2019] [Indexed: 11/16/2022] Open
Abstract
Alterations in circadian rhythms are closely linked to depression, and we have shown earlier that progressive alterations in circadian entrainment precede the onset of depression in mice exposed in utero to excess glucocorticoids. The aim of this study was to investigate whether treatment with the noradrenaline reuptake inhibitor desipramine (DMI) could restore the alterations in circadian entrainment and prevent the onset of depression-like behavior. C57Bl/6 mice were exposed to dexamethasone (DEX-synthetic glucocorticoid analog, 0.05 mg/kg/day) between gestational day 14 and delivery. Male offspring aged 6 months (mo) were treated with DMI (10 mg/kg/day in drinking water) for at least 21 days before behavioral testing. We recorded spontaneous activity using the TraffiCage™ system and found that DEX mice re-entrained faster than controls after an abrupt advance in light-dark cycle by 6 h, while DMI treatment significantly delayed re-entrainment. Next we assessed the synchronization of peripheral oscillators with the central clock (located in the suprachiasmatic nucleus-SCN), as well as the mechanisms required for entrainment. We found that photic entrainment of the SCN was apparently preserved in DEX mice, but the expression of clock genes in the hippocampus was not synchronized with the light-dark cycle. This was associated with downregulated mRNA expression for arginine vasopressin (AVP; the main molecular output entraining peripheral clocks) in the SCN, and for glucocorticoid receptor (GR; required for the negative feedback loop regulating glucocorticoid secretion) in the hippocampus. DMI treatment restored the mRNA expression of AVP in the SCN and enhanced GR-mediated signaling by upregulating GR expression and nuclear translocation in the hippocampus. Furthermore, DMI treatment at 6 mo prevented the onset of depression-like behavior and the associated alterations in neurogenesis in 12-mo-old DEX mice. Taken together, our data indicate that DMI treatment enhances GR-mediated signaling and restores the synchronization of peripheral clocks with the SCN and support the hypothesis that altered circadian entrainment is a modifiable risk factor for depression.
Collapse
Affiliation(s)
- Stefan Spulber
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Mirko Conti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Marilena Raciti
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Sandra Ceccatelli
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
48
|
Manjur ABMK, Lempiäinen JK, Malinen M, Palvimo JJ, Niskanen EA. IRF2BP2 modulates the crosstalk between glucocorticoid and TNF signaling. J Steroid Biochem Mol Biol 2019; 192:105382. [PMID: 31145973 DOI: 10.1016/j.jsbmb.2019.105382] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
IRF2BP2 (interferon regulatory factor-2 binding protein-2) is an uncharacterized interaction partner of glucocorticoid (GC) receptor (GR), an anti-inflammatory and metabolic transcription factor. Here, we show that GC changes the chromatin binding of IRF2BP2 in natural chromatin milieu. The GC-induced IRF2BP2-binding sites co-occur with GR binding sites and are associated with GC-induced genes. Moreover, the depletion of IRF2BP2 modulates transcription of GC-regulated genes, represses cell proliferation and increases cell movement in HEK293 cells. In A549 cells, the depletion extensively alters the responses to GC and tumor necrosis factor α (TNF), including metabolic and inflammatory pathways. Taken together, our data support the role of IRF2BP2 as a coregulator of both GR and NF-κB, potentially modulating the crosstalk between GC and TNF signaling.
Collapse
Affiliation(s)
| | | | - Marjo Malinen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland; Department of Environmental and Biological Sciences, University of Eastern Finland, Joensuu, Finland
| | - Jorma J Palvimo
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| | - Einari A Niskanen
- Institute of Biomedicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
49
|
Differential transcriptional response following glucocorticoid activation in cultured blood immune cells: a novel approach to PTSD biomarker development. Transl Psychiatry 2019; 9:201. [PMID: 31434874 PMCID: PMC6704073 DOI: 10.1038/s41398-019-0539-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/23/2019] [Accepted: 07/07/2019] [Indexed: 12/21/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) is a condition of stress reactivity, whose clinical manifestations are evident when patients are triggered following exposure to a traumatic event. While baseline differences in gene expression of glucocorticoid signaling and inflammatory cytokines in peripheral blood mononuclear cells (PBMCs) have been associated with PTSD, these alterations do not fully recapitulate the molecular response to physiological triggers, such as stress hormones. Therefore, it is critical to develop new techniques that will capture the dynamic transcriptional response associated with stress-activated conditions relative to baseline conditions. To achieve this goal, cultured PBMCs from combat-exposed veterans with PTSD(+) (n = 10) and without PTSD(-) (n = 10) were incubated with increasing concentrations (vehicle, 2.5 nM, 5 nM, 50 nM) of dexamethasone (DEX). Across diagnosis and dosage, several genes and gene networks were reliable markers of glucocorticoid stimulation (FDR < 5%), including enhanced expression of FKPB5, VIPR1, NR1I3, and apoptosis-related pathways, and reduced expression of NR3C1, STAT1, IRF1, and related inflammatory and cellular stress-responsive pathways. Dose-dependent differential transcriptional changes in several genes were also identified between PTSD+ and PTSD-. Robust changes in expression were observed at 2.5 nM DEX in PTSD- but not PTSD+ participants; whereas, with increasing concentrations (5 nM and 50 nM), several genes were identified to be uniquely up-regulated in PTSD+ but not PTSD- participants. Collectively, these preliminary findings suggest that genome-wide gene expression profiling of DEX-stimulated PBMCs is a promising method for the exploration of the dynamic differential molecular responses to stress hormones in PTSD, and may identify novel markers of altered glucocorticoid signaling and responsivity in PTSD.
Collapse
|
50
|
Strategies to Overcome Resistance Mechanisms in T-Cell Acute Lymphoblastic Leukemia. Int J Mol Sci 2019; 20:ijms20123021. [PMID: 31226848 PMCID: PMC6627878 DOI: 10.3390/ijms20123021] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/14/2019] [Accepted: 06/17/2019] [Indexed: 12/20/2022] Open
Abstract
Chemoresistance is a major cause of recurrence and death from T-cell acute lymphoblastic leukemia (T-ALL), both in adult and pediatric patients. In the majority of cases, drug-resistant disease is treated by selecting a combination of other drugs, without understanding the molecular mechanisms by which malignant cells escape chemotherapeutic treatments, even though a more detailed genomic characterization and the identification of actionable disease targets may enable informed decision of new agents to improve patient outcomes. In this work, we describe pathways of resistance to common chemotherapeutic agents including glucocorticoids and review the resistance mechanisms to targeted therapy such as IL7R, PI3K-AKT-mTOR, NOTCH1, BRD4/MYC, Cyclin D3: CDK4/CDK6, BCL2 inhibitors, and selective inhibitors of nuclear export (SINE). Finally, to overcome the limitations of the current trial-and-error method, we summarize the experiences of anti-cancer drug sensitivity resistance profiling (DSRP) approaches as a rapid and relevant strategy to infer drug activity and provide functional information to assist clinical decision one patient at a time.
Collapse
|