51
|
Inflammatory macrophage memory in nonsteroidal anti-inflammatory drug-exacerbated respiratory disease. J Allergy Clin Immunol 2020; 147:587-599. [PMID: 32540397 DOI: 10.1016/j.jaci.2020.04.064] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/04/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Nonsteroidal anti-inflammatory drug-exacerbated respiratory disease (N-ERD) is a chronic inflammatory condition, which is driven by an aberrant arachidonic acid metabolism. Macrophages are major producers of arachidonic acid metabolites and subject to metabolic reprogramming, but they have been neglected in N-ERD. OBJECTIVE This study sought to elucidate a potential metabolic and epigenetic macrophage reprogramming in N-ERD. METHODS Transcriptional, metabolic, and lipid mediator profiles in macrophages from patients with N-ERD and healthy controls were assessed by RNA sequencing, Seahorse assays, and LC-MS/MS. Metabolites in nasal lining fluid, sputum, and plasma from patients with N-ERD (n = 15) and healthy individuals (n = 10) were quantified by targeted metabolomics analyses. Genome-wide methylomics were deployed to define epigenetic mechanisms of macrophage reprogramming in N-ERD. RESULTS This study shows that N-ERD monocytes/macrophages exhibit an overall reduction in DNA methylation, aberrant metabolic profiles, and an increased expression of chemokines, indicative of a persistent proinflammatory activation. Differentially methylated regions in N-ERD macrophages included genes involved in chemokine signaling and acylcarnitine metabolism. Acylcarnitines were increased in macrophages, sputum, nasal lining fluid, and plasma of patients with N-ERD. On inflammatory challenge, N-ERD macrophages produced increased levels of acylcarnitines, proinflammatory arachidonic acid metabolites, cytokines, and chemokines as compared to healthy macrophages. CONCLUSIONS Together, these findings decipher a proinflammatory metabolic and epigenetic reprogramming of macrophages in N-ERD.
Collapse
|
52
|
Effect of congenital adrenal hyperplasia treated by glucocorticoids on plasma metabolome: a machine-learning-based analysis. Sci Rep 2020; 10:8859. [PMID: 32483270 PMCID: PMC7264133 DOI: 10.1038/s41598-020-65897-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 05/11/2020] [Indexed: 12/30/2022] Open
Abstract
Background. Congenital adrenal hyperplasia (CAH) due to 21-hydroxylase deficiency leads to impaired cortisol biosynthesis. Treatment includes glucocorticoid supplementation. We studied the specific metabolomics signatures in CAH patients using two different algorithms. Methods. In a case-control study of CAH patients matched on sex and age with healthy control subjects, two metabolomic analyses were performed: one using MetaboDiff, a validated differential metabolomic analysis tool and the other, using Predomics, a novel machine-learning algorithm. Results. 168 participants were included (84 CAH patients). There was no correlation between plasma cortisol levels during glucocorticoid supplementation and metabolites in CAH patients. Indoleamine 2,3-dioxygenase enzyme activity was correlated with ACTH (rho coefficient = −0.25, p-value = 0.02), in CAH patients but not in controls subjects. Overall, 33 metabolites were significantly altered in CAH patients. Main changes came from: purine and pyrimidine metabolites, branched aminoacids, tricarboxylic acid cycle metabolites and associated pathways (urea, glucose, pentose phosphates). MetaboDiff identified 2 modules that were significantly different between both groups: aminosugar metabolism and purine metabolism. Predomics found several interpretable models which accurately discriminated the two groups (accuracy of 0.86 and AUROC of 0.9). Conclusion. CAH patients and healthy control subjects exhibit significant differences in plasma metabolomes, which may be explained by glucocorticoid supplementation.
Collapse
|
53
|
Dexamethasone-Induced Perturbations in Tissue Metabolomics Revealed by Chemical Isotope Labeling LC-MS analysis. Metabolites 2020; 10:metabo10020042. [PMID: 31973046 PMCID: PMC7074358 DOI: 10.3390/metabo10020042] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/19/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022] Open
Abstract
Dexamethasone (Dex) is a synthetic glucocorticoid (GC) drug commonly used clinically for the treatment of several inflammatory and immune-mediated diseases. Despite its broad range of indications, the long-term use of Dex is known to be associated with specific abnormalities in several tissues and organs. In this study, the metabolomic effects on five different organs induced by the chronic administration of Dex in the Sprague–Dawley rat model were investigated using the chemical isotope labeling liquid chromatography-mass spectrometry (CIL LC-MS) platform, which targets the amine/phenol submetabolomes. Compared to controls, a prolonged intake of Dex resulted in significant perturbations in the levels of 492, 442, 300, 186, and 105 metabolites in the brain, skeletal muscle, liver, kidney, and heart tissues, respectively. The positively identified metabolites were mapped to diverse molecular pathways in different organs. In the brain, perturbations in protein biosynthesis, amino acid metabolism, and monoamine neurotransmitter synthesis were identified, while in the heart, pyrimidine metabolism and branched amino acid biosynthesis were the most significantly impaired pathways. In the kidney, several amino acid pathways were dysregulated, which reflected impairments in several biological functions, including gluconeogenesis and ureagenesis. Beta-alanine metabolism and uridine homeostasis were profoundly affected in liver tissues, whereas alterations of glutathione, arginine, glutamine, and nitrogen metabolism pointed to the modulation of muscle metabolism and disturbances in energy production and muscle mass in skeletal muscle. The differential expression of multiple dipeptides was most significant in the liver (down-regulated), brain (up-regulation), and kidney tissues, but not in the heart or skeletal muscle tissues. The identification of clinically relevant pathways provides holistic insights into the tissue molecular responses induced by Dex and understanding of the underlying mechanisms associated with their side effects. Our data suggest a potential role for glutathione supplementation and dipeptide modulators as novel therapeutic interventions to mitigate the side effects induced by Dex therapy.
Collapse
|
54
|
Dey KK, Ghosh M. Understanding the structure and dynamics of anti-inflammatory corticosteroid dexamethasone by solid state NMR spectroscopy. RSC Adv 2020; 10:37564-37575. [PMID: 35521289 PMCID: PMC9057149 DOI: 10.1039/d0ra05474g] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 10/01/2020] [Indexed: 12/31/2022] Open
Abstract
For decades corticosteroid dexamethasone has been applied as an anti-inflammatory, immunosuppressant, and decongestant, in the prevention of postoperative nausea and vomiting (PONV), and for auto-immune diseases, allergic reactions, total hip arthroplasty (THA), and cancer. Recently in vitro studies suggested that it may be beneficial to deal with the COVID-19 pandemic. This important drug molecule was investigated by solid state NMR measurements to provide more complete features of its structure and dynamics at atomic scale resolution. The spin–lattice relaxation time at twenty-two different carbon sites of dexamethasone was determined by the Torchia CP method. The principle components of the chemical shift anisotropy tensor were determined by 13C two-dimensional phase adjusted spinning sideband (2DPASS) cross-polarization magic angle spinning (CP-MAS) solid state NMR experiments. The molecular correlation time at twenty-two crystallographically different carbon sites of dexamethasone was calculated by considering that the spin–lattice relaxation mechanism of the 13C nucleus is mainly governed by the chemical shift anisotropy interaction and the heteronuclear dipole–dipole coupling. The spin–lattice relaxation time of carbon nuclei resides on ‘A’, ‘B’, ‘C’, and ‘D’ rings and the side-chain of dexamethasone is quite large, which implies the close-packed arrangement of the molecule. The difference in molecular correlation time at various regions of the molecule demonstrates the existence of different degrees of freedom within the molecule. This may be the reason for the various biological activities exhibited by the molecule. These types of detailed features of the structure and dynamics of such an important drug with multiple biological activities are necessary to develop the advanced medicine and it will also help to understand the structure–activity relationships of corticosteroid. The structure and dynamics of dexamethasone is determined by measuring CSA tensor, site-specific spin–lattice relaxation time.![]()
Collapse
Affiliation(s)
- Krishna Kishor Dey
- Department of Physics
- Dr. Harisingh Gour Central University
- Sagar-470003
- India
| | - Manasi Ghosh
- Physics Section
- MMV
- Banaras Hindu University
- Varanasi-221005
- India
| |
Collapse
|
55
|
Cordani M, Strippoli R, Somoza Á. Nanomaterials as Inhibitors of Epithelial Mesenchymal Transition in Cancer Treatment. Cancers (Basel) 2019; 12:E25. [PMID: 31861725 PMCID: PMC7017008 DOI: 10.3390/cancers12010025] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/13/2019] [Indexed: 02/06/2023] Open
Abstract
Abstract: Epithelial-mesenchymal transition (EMT) has emerged as a key regulator of cell invasion and metastasis in cancers. Besides the acquisition of migratory/invasive abilities, the EMT process is tightly connected with the generation of cancer stem cells (CSCs), thus contributing to chemoresistance. However, although EMT represents a relevant therapeutic target for cancer treatment, its application in the clinic is still limited due to various reasons, including tumor-stage heterogeneity, molecular-cellular target specificity, and appropriate drug delivery. Concerning this last point, different nanomaterials may be used to counteract EMT induction, providing novel therapeutic tools against many different cancers. In this review, (1) we discuss the application of various nanomaterials for EMT-based therapies in cancer, (2) we summarize the therapeutic relevance of some of the proposed EMT targets, and (3) we review the potential benefits and weaknesses of each approach.
Collapse
Affiliation(s)
- Marco Cordani
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain
| | - Raffaele Strippoli
- Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy;
- National Institute for Infectious Diseases “Lazzaro Spallanzani” I.R.C.C.S., 00149 Rome, Italy
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain
- CNB-CSIC-IMDEA Nanociencia Associated Unit “Unidad de Nanobiotecnología”, 28049 Madrid, Spain
| |
Collapse
|
56
|
Abstract
While glucocorticoids have been used for over 50 years to treat rheumatoid and osteoarthritis pain, the prescription of glucocorticoids remains controversial because of potentially harmful side effects at the molecular, cellular and tissue levels. One member of the glucocorticoid family, dexamethasone (DEX) has recently been demonstrated to rescue cartilage matrix loss and chondrocyte viability in animal studies and cartilage explant models of tissue injury and post-traumatic osteoarthritis, suggesting the possibility of DEX as a disease-modifying drug if used appropriately. However, the literature on the effects of DEX on cartilage reveals conflicting results on the drug's safety, depending on the dose and duration of DEX exposure as well as the model system used. Overall, DEX has been shown to protect against arthritis-related changes in cartilage structure and function, including matrix loss, inflammation and cartilage viability. These beneficial effects are not always observed in model systems using initially healthy cartilage or isolated chondrocytes, where many studies have reported significant increases in chondrocyte apoptosis. It is crucially important to understand under what conditions DEX may be beneficial or harmful to cartilage and other joint tissues and to determine potential for safe use of this glucocorticoid in the clinic as a disease-modifying drug.
Collapse
Affiliation(s)
- R. Black
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - A. J. Grodzinsky
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA,Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA,Address for correspondence: Prof. Al Grodzinsky, MIT, Centre for Biomedical Engineering, 500 Technology Square, Cambridge, MA, 02139, USA.
| |
Collapse
|
57
|
Phytochemical Investigation of Tradescantia Albiflora and Anti-Inflammatory Butenolide Derivatives. Molecules 2019; 24:molecules24183336. [PMID: 31540241 PMCID: PMC6767271 DOI: 10.3390/molecules24183336] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022] Open
Abstract
Phytochemical investigation of the whole plant of Tradescantia albiflora Kunth led to the isolation and characterization of a butanolide, rosmarinosin B (1), that was isolated from natural sources for the first time, a new butenolide, 5-O-acetyl bracteanolide A (2), and a new apocarotenoid, 2β-hydroxyisololiolide (11), together with 25 known compounds (compounds 3–10 and 12–28). The structures of the new compounds were elucidated by analysis of their spectroscopic data, including MS, 1D, and 2D NMR experiments, and comparison with literature data of known compounds. Furthermore, four butenolides 4a–4d were synthesized as novel derivatives of bracteanolide A. The isolates and the synthesized derivatives were evaluated for their preliminary anti-inflammatory activity against lipopolysaccharide (LPS)-stimulated nitric oxide (NO) production in RAW 264.7 cells. Among them, the synthesized butenolide derivative n-butyl bracteanolide A (4d) showed enhanced NO inhibitory activity compared to the original compound, with an IC50 value of 4.32 ± 0.09 μg/mL.
Collapse
|
58
|
Kiernozek E, Bieńkowska A, Markowska M, Kozlowska E, Drela N. Dexamethasone affects day/night development and function of thymus-derived T regulatory cells. Immunobiology 2019; 224:614-624. [PMID: 31427114 DOI: 10.1016/j.imbio.2019.07.007] [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: 05/24/2019] [Accepted: 07/30/2019] [Indexed: 11/19/2022]
Abstract
Thymus-derived T regulatory (tTregs) cells play a crucial role in the maintenance of tolerance and immune homeostasis. Mechanisms and factors regulating tTreg development and function are widely investigated, but to a large degree still remain unclear. Our previous findings demonstrated that, in physiological conditions, the development and suppressive function of tTregs demonstrated day/night rhythmicity, which correlated with the concentration of plasma corticosterone and the expression of glucocorticoid receptors. In this study we ask whether synthetic glucocorticoids commonly used to inhibit excessive activity of the immune system, can modulate the development and suppressive function of tTregs in vivo depending on the time of administration. Young C57BL/6 male and female mice were injected intraperitoneally with a single dose of dexamethasone at two time points of the day: 7.00-8.00 a.m. and 7.00-8.00 p.m. The experimental can be used to indicate on the potentially expected positive or adverse side effects and can constitute also a good model for the assessment of the effects of long-term therapy. The results of our studies demonstrated the increase of the percentage of tTregs at both time points in male mice, but only in the evening in females. The suppressive activity of tTregs increased independently on the day time of in female mice, but in the morning only in males. We concluded that in the condition of dexamethasone supplementation, the elevated suppressive potential of tTregs is balanced by the induction apoptosis in order to prevent excessive suppression.
Collapse
Affiliation(s)
- Ewelina Kiernozek
- Faculty of Biology, University of Warsaw, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Anna Bieńkowska
- Faculty of Biology, University of Warsaw, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Magdalena Markowska
- Faculty of Biology, University of Warsaw, Department of Animal Physiology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Ewa Kozlowska
- Faculty of Biology, University of Warsaw, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland
| | - Nadzieja Drela
- Faculty of Biology, University of Warsaw, Department of Immunology, Miecznikowa 1, 02-096 Warsaw, Poland.
| |
Collapse
|
59
|
Malkawi AK, Masood A, Shinwari Z, Jacob M, Benabdelkamel H, Matic G, Almuhanna F, Dasouki M, Alaiya AA, Rahman AMA. Proteomic Analysis of Morphologically Changed Tissues after Prolonged Dexamethasone Treatment. Int J Mol Sci 2019; 20:ijms20133122. [PMID: 31247941 PMCID: PMC6650964 DOI: 10.3390/ijms20133122] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/16/2019] [Accepted: 06/18/2019] [Indexed: 12/16/2022] Open
Abstract
Prolonged dexamethasone (Dex) administration leads to serious adverse and decrease brain and heart size, muscular atrophy, hemorrhagic liver, and presence of kidney cysts. Herein, we used an untargeted proteomic approach using liquid chromatography-tandem mass spectrometry (LC-MS/MS) for simultaneous identification of changes in proteomes of the major organs in Sprague–Dawley (SD rats post Dex treatment. The comparative and quantitative proteomic analysis of the brain, heart, muscle, liver, and kidney tissues revealed differential expression of proteins (n = 190, 193, 39, 230, and 53, respectively) between Dex-treated and control rats. Functional network analysis using ingenuity pathway analysis (IPA revealed significant differences in regulation of metabolic pathways within the morphologically changed organs that related to: (i) brain—cell morphology, nervous system development, and function and neurological disease; (ii) heart—cellular development, cellular function and maintenance, connective tissue development and function; (iii) skeletal muscle—nucleic acid metabolism, and small molecule biochemical pathways; (iv) liver—lipid metabolism, small molecular biochemistry, and nucleic acid metabolism; and (v) kidney—drug metabolism, organism injury and abnormalities, and renal damage. Our study provides a comprehensive description of the organ-specific proteomic profilesand differentially altered biochemical pathways, after prolonged Dex treatement to understand the molecular basis for development of side effects.
Collapse
Affiliation(s)
- Abeer K Malkawi
- Department of Chemistry and Biochemistry, Concordia University, 7141 Sherbrook Street West, Montréal, QC H4B 1R6, Canada
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia
| | - Zakia Shinwari
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia
| | - Minnie Jacob
- Department of Genetics, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia
- College of Public Health, Medical, and Veterinary Sciences/Molecular & Cell Biology, James Cook University, Townsville, QLD 4811, Australia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, P.O. Box 2925 (98), Riyadh 11461, Saudi Arabia
| | - Goran Matic
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia
| | - Falah Almuhanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia
| | - Majed Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia
| | - Ayodele A Alaiya
- Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia
| | - Anas M Abdel Rahman
- Department of Genetics, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh 11461, Saudi Arabia.
- College of Medicine, Al Faisal University, Riyadh 11533, Saudi Arabia.
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL A1B 3X7, Canada.
| |
Collapse
|
60
|
Meka RR, Venkatesha SH, Acharya B, Moudgil KD. Peptide-targeted liposomal delivery of dexamethasone for arthritis therapy. Nanomedicine (Lond) 2019; 14:1455-1469. [PMID: 30938236 PMCID: PMC6613046 DOI: 10.2217/nnm-2018-0501] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
Aim: Rheumatoid arthritis is an autoimmune disease affecting the joints. Antiarthritic drugs are given systemically, thereby exposing various healthy organs to these drugs, resulting in adverse reactions. Accordingly, there is an urgent need for targeted drug delivery methods for inflamed joints. Materials & methods: We developed a liposomal drug delivery system using a novel peptide ligand (CKPFDRALC) named ART-2, which homes to the inflamed joints when injected intravenously to rats with adjuvant-induced arthritis. Results: The ART-2-coated liposomes encapsulating an antiarthritic drug, dexamethasone (DEX), were more effective in inhibiting arthritis progression than control-DEX liposomes or free DEX, despite a comparable safety profile. Conclusion: Peptide-targeted therapy has advantages over conventional drug delivery and can be adapted for rheumatoid arthritis therapy.
Collapse
Affiliation(s)
- Rakeshchandra R Meka
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shivaprasad H Venkatesha
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Bodhraj Acharya
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kamal D Moudgil
- Baltimore Veterans Affairs Medical Center, Baltimore, MD 21201, USA
- Department of Microbiology & Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Department of Medicine, Division of Rheumatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
61
|
Changes in the Canine Plasma Lipidome after Short- and Long-Term Excess Glucocorticoid Exposure. Sci Rep 2019; 9:6015. [PMID: 30979907 PMCID: PMC6461633 DOI: 10.1038/s41598-019-42190-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 03/20/2019] [Indexed: 02/07/2023] Open
Abstract
Glucocorticoids (GCs) are critical regulators of metabolic control in mammals and their aberrant function has been linked to several pathologies. GCs are widely used in human and veterinary clinical practice as potent anti-inflammatory and immune suppressive agents. Dyslipidaemia is a frequently observed consequence of GC treatment, typified by increased lipolysis, lipid mobilization, liponeogenesis, and adipogenesis. Dogs with excess GC show hyperlipidaemia, hypertension, and a higher risk of developing type 2 diabetes mellitus, but the risk of developing atherosclerotic lesions is low as compared to humans. This study aimed to examine alterations in the canine plasma lipidome in a model of experimentally induced short-term and long-term GC excess. Both treatments led to significant plasma lipidome alterations, which were more pronounced after long-term excess steroid exposure. In particular, monohexosylceramides, phosphatidylinositols, ether phosphatidylcholines, acyl phosphatidylcholines, triacylglycerols and sphingosine 1-phosphates showed significant changes. The present study highlights the hitherto unknown effects of GCs on lipid metabolism, which will be important in the further elucidation of the role and function of GCs as drugs and in metabolic and cardiovascular diseases.
Collapse
|
62
|
Kusuda Y, Kondo Y, Miyagi Y, Munemasa T, Hori Y, Aonuma F, Tsuka S, Mukaibo T, Masaki C, Hosokawa R. Long-term dexamethasone treatment diminishes store-operated Ca 2+ entry in salivary acinar cells. Int J Oral Sci 2019; 11:1. [PMID: 30602784 PMCID: PMC6315037 DOI: 10.1038/s41368-018-0031-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 11/09/2022] Open
Abstract
Corticosteroids are used in the treatment of many diseases; however, they also induce various side effects. Dexamethasone is one of the most potent corticosteroids, and it has been reported to induce the side effect of impaired salivary gland function. This study aimed to evaluate the effects of dexamethasone on mouse submandibular gland function to gain insight into the mechanism of dexamethasone-induced salivary hypofunction. The muscarinic agonist carbachol (CCh) induced salivary secretion and was not affected by short-term dexamethasone treatment but was decreased following long-term dexamethasone administration. The expression levels of the membrane proteins Na+-K+-2Cl- cotransporter, transmembrane member 16A, and aquaporin 5 were comparable between the control and long-term dexamethasone treatment groups. The CCh-induced increase in calcium concentration was significantly lower in the presence of extracellular Ca2+ in the long-term dexamethasone treatment group compared to that in the control group. Furthermore, CCh-induced salivation in the absence of extracellular Ca2+ and Ca2+ ionophore A23187-induced salivation was comparable between the control and long-term dexamethasone treatment groups. Moreover, salivation induced by the Ca2+-ATPase inhibitor thapsigargin was diminished in the long-term dexamethasone treatment group. In summary, these results demonstrate that short-term dexamethasone treatment did not impair salivary gland function, whereas long-term dexamethasone treatment diminished store-operated Ca2+ entry, resulting in hyposalivation in mouse submandibular glands.
Collapse
Affiliation(s)
- Yuichiro Kusuda
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Yusuke Kondo
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan.
| | - Yuta Miyagi
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Takashi Munemasa
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Yusuke Hori
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Fumiko Aonuma
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Shintaro Tsuka
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Taro Mukaibo
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Chihiro Masaki
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Ryuji Hosokawa
- Division of Oral Reconstruction and Rehabilitation, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| |
Collapse
|
63
|
Zhu B, Wang L, Huang J, Xiang X, Tang Y, Cheng C, Yan F, Ma L, Qiu L. Ultrasound-triggered perfluorocarbon-derived nanobombs for targeted therapies of rheumatoid arthritis. J Mater Chem B 2019. [DOI: 10.1039/c9tb00978g] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The targeted US-triggered PFC-based “nanobombs” with US used to treat the RA in this work would offer a new treatment strategy and have a great potential for the application in the areas of theranostic agent and nanomedicine treatment.
Collapse
Affiliation(s)
- Bihui Zhu
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Liyun Wang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Jianbo Huang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Xi Xiang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Yuanjiao Tang
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Chong Cheng
- College of Polymer Science and Engineering
- State Key Laboratory of Polymer Materials Engineering
- Sichuan University
- Chengdu 610065
- P. R. China
| | - Feng Yan
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Lang Ma
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| | - Li Qiu
- Department of Ultrasound
- Laboratory of Ultrasound Imaging Drug
- West China Hospital
- Sichuan University
- Chengdu 610041
| |
Collapse
|
64
|
Lee CH. Epithelial-mesenchymal transition: Initiation by cues from chronic inflammatory tumor microenvironment and termination by anti-inflammatory compounds and specialized pro-resolving lipids. Biochem Pharmacol 2018; 158:261-273. [DOI: 10.1016/j.bcp.2018.10.031] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 10/29/2018] [Indexed: 02/07/2023]
|
65
|
Zhang X, Geven MA, Wang X, Qin L, Grijpma DW, Peijs T, Eglin D, Guillaume O, Gautrot JE. A drug eluting poly(trimethylene carbonate)/poly(lactic acid)-reinforced nanocomposite for the functional delivery of osteogenic molecules. Int J Nanomedicine 2018; 13:5701-5718. [PMID: 30288042 PMCID: PMC6161751 DOI: 10.2147/ijn.s163219] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Background Poly(trimethylene carbonate) (PTMC) has wide biomedical applications in the field of tissue engineering, due to its biocompatibility and biodegradability features. Its common manufacturing involves photofabrication, such as stereolithography (SLA), which allows the fabrication of complex and controlled structures. Despite the great potential of SLA-fabricated scaffolds, very few examples of PTMC-based drug delivery systems fabricated using photo-fabrication can be found ascribed to light-triggered therapeutics instability, degradation, side reaction, binding to the macromers, etc. These concerns severely restrict the development of SLA-fabricated PTMC structures for drug delivery purposes. Methods In this context, we propose here, as a proof of concept, to load a drug model (dexamethasone) into electrospun fibers of poly(lactic acid), and then to integrate these bioactive fibers into the photo-crosslinkable resin of PTMC to produce hybrid films. The hybrid films' properties and drug release profile were characterized; its biological activity was investigated via bone marrow mesenchymal stem cells culture and differentiation assays. Results The polymer/polymer hybrids exhibit improved properties compared with PTMC-only films, in terms of mechanical performance and drug protection from UV denaturation. We further validated that the dexamethasone preserved its biological activity even after photoreaction within the PTMC/poly(lactic acid) hybrid structures by investigating bone marrow mesenchymal stem cells proliferation and osteogenic differentiation. Conclusion This study demonstrates the potential of polymer-polymer scaffolds to simultaneously reinforce the mechanical properties of soft matrices and to load sensitive drugs in scaffolds that can be fabricated via additive manufacturing.
Collapse
Affiliation(s)
- Xi Zhang
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, UK, .,Institute of Bioengineering, Queen Mary University of London, Mile End Road, London, UK,
| | - Mike A Geven
- Department of Biomaterials Science and Technology, University of Twente, Enschede, the Netherlands
| | - Xinluan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 5018057, China
| | - Ling Qin
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 5018057, China
| | - Dirk W Grijpma
- Department of Biomaterials Science and Technology, University of Twente, Enschede, the Netherlands
| | - Ton Peijs
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, UK,
| | - David Eglin
- AO Research Institute Davos, Davos, Switzerland,
| | | | - Julien E Gautrot
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London, UK, .,Institute of Bioengineering, Queen Mary University of London, Mile End Road, London, UK,
| |
Collapse
|
66
|
Barghi K, Edmonds KP, Ajayi TA, Atayee RS. Prescribing Trends of Palliative Care Team's Use of Dexamethasone for Cancer-Related Pain. J Pain Palliat Care Pharmacother 2018; 32:37-43. [PMID: 29737903 DOI: 10.1080/15360288.2018.1460436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Opioids are first-line therapy for cancer-related pain. In addition, corticosteroids are commonly utilized as adjuvant analgesics for pain and other symptoms in the oncology setting with limited supporting data. A retrospective analysis was conducted evaluating adult hospitalized patients receiving opioids who received once-daily dexamethasone on the recommendation of a specialty palliative care team during their hospitalization from January 1, 2015, to January 1, 2016. Primary end point was to describe prescribing patterns of dexamethasone in this patient population and secondarily examining any effect on oral morphine equivalent daily dose (MEDD), numeric pain score (NPS), and unwanted effects at 24 and 48 hours after the first dose of dexamethasone. Fifty-nine patients received an average dose of 13 mg (SD = 10) of dexamethasone for cancer-related pain, primarily acute pain (n = 36, 61%). Many died before hospital discharge or soon thereafter (n = 28, 47.5%). Although not statistically significant, our study shows a decrease of 23% and 19% in MEDD and NPS, respectively, without change in WBC after dexamethasone. A specialty palliative care team most often used once-daily dexamethasone for cancer-related pain in patients near the end of life. There were trends toward lower MEDD and NPS, but more robust studies are needed for validation.
Collapse
|
67
|
Malkawi AK, Alzoubi KH, Jacob M, Matic G, Ali A, Al Faraj A, Almuhanna F, Dasouki M, Abdel Rahman AM. Metabolomics Based Profiling of Dexamethasone Side Effects in Rats. Front Pharmacol 2018; 9:46. [PMID: 29503615 PMCID: PMC5820529 DOI: 10.3389/fphar.2018.00046] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/15/2018] [Indexed: 01/05/2023] Open
Abstract
Dexamethasone (Dex) is a synthetic glucocorticoid that has anti-inflammatory and immunosuppressant effects and is used in several conditions such as asthma and severe allergy. Patients receiving Dex, either at a high dose or for a long time, might develop several side effects such as hyperglycemia, weight change, or osteoporosis due to its in vivo non-selectivity. Herein, we used liquid chromatography-tandem mass spectrometry-based comprehensive targeted metabolomic profiling as well as radiographic imaging techniques to study the side effects of Dex treatment in rats. The Dex-treated rats suffered from a ∼20% reduction in weight gain, hyperglycemia (145 mg/dL), changes in serum lipids, and reduction in total serum alkaline phosphatase (ALP) (∼600 IU/L). Also, compared to controls, Dex-treated rats showed a distinctive metabolomics profile. In particular, serum amino acids metabolism showed six-fold reduction in phenylalanine, lysine, and arginine levels and upregulation of tyrosine and hydroxyproline reflecting perturbations in gluconeogenesis and protein catabolism which together lead to weight loss and abnormal bone metabolism. Sorbitol level was markedly elevated secondary to hyperglycemia and reflecting activation of the polyol metabolism pathway causing a decrease in the availability of reducing molecules (glutathione, NADPH, NAD+). Overexpression of succinylacetone (4,6-dioxoheptanoic acid) suggests a novel inhibitory effect of Dex on hepatic fumarylacetoacetate hydrolase. The acylcarnitines, mainly the very long chain species (C12, C14:1, C18:1) were significantly increased after Dex treatment which reflects degradation of the adipose tissue. In conclusion, long-term Dex therapy in rats is associated with a distinctive metabolic profile which correlates with its side effects. Therefore, metabolomics based profiling may predict Dex treatment-related side effects and may offer possible novel therapeutic interventions.
Collapse
Affiliation(s)
- Abeer K Malkawi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Karem H Alzoubi
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Minnie Jacob
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- Molecular and Cell Biology, College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, QLD, Australia
| | - Goran Matic
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Asmaa Ali
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Achraf Al Faraj
- Department of Radiologic Sciences, Faculty of Health Sciences, American University of Science and Technology, Beirut, Lebanon
| | - Falah Almuhanna
- Department of Comparative Medicine, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Majed Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Anas M Abdel Rahman
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, Canada
| |
Collapse
|
68
|
Design, synthesis, and biological evaluation of novel catecholopyrimidine based PDE4 inhibitor for the treatment of atopic dermatitis. Eur J Med Chem 2018; 145:673-690. [DOI: 10.1016/j.ejmech.2017.12.069] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/19/2022]
|
69
|
Alwashih MA, Stimson RH, Andrew R, Walker BR, Watson DG. Acute interaction between hydrocortisone and insulin alters the plasma metabolome in humans. Sci Rep 2017; 7:11488. [PMID: 28904371 PMCID: PMC5597623 DOI: 10.1038/s41598-017-10200-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 08/07/2017] [Indexed: 12/30/2022] Open
Abstract
With the aim of identifying biomarkers of glucocorticoid action and their relationship with biomarkers of insulin action, metabolomic profiling was carried out in plasma samples from twenty healthy men who were administered either a low or medium dose insulin infusion (n = 10 each group). In addition, all subjects were given metyrapone (to inhibit adrenal cortisol secretion) + /− hydrocortisone (HC) in a randomised crossover design to produce low, medium and high glucocorticoid levels. The clearest effects of insulin were to reduce plasma levels of the branched chain amino acids (BCAs) leucine/isoleucine and their deaminated metabolites, and lowered free fatty acids and acylcarnitines. The highest dose of hydrocortisone increased plasma BCAs in both insulin groups but increased free fatty acids only in the high insulin group, however hydrocortisone did not affect the levels of acyl carnitines in either group. The clearest interaction between HC and insulin was that hydrocortisone produced an elevation in levels of BCAs and their metabolites which were lowered by insulin. The direct modulation of BCAs by glucocorticoids and insulin may provide the basis for improved in vivo monitoring of glucocorticoid and insulin action.
Collapse
Affiliation(s)
- Mohammad A Alwashih
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK.,General Directorate of Medical Services, Ministry of Interior, Riyadh, 13321, Saudi Arabia
| | - Roland H Stimson
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Scotland, UK
| | - Ruth Andrew
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Scotland, UK
| | - Brian R Walker
- BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, University of Edinburgh, Scotland, UK
| | - David G Watson
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK.
| |
Collapse
|
70
|
Large Scale Metabolic Profiling identifies Novel Steroids linked to Rheumatoid Arthritis. Sci Rep 2017; 7:9137. [PMID: 28831053 PMCID: PMC5567269 DOI: 10.1038/s41598-017-05439-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 06/01/2017] [Indexed: 01/06/2023] Open
Abstract
Recent metabolomics studies of Rheumatoid Arthritis (RA) reported few metabolites that were associated with the disease, either due to small cohort sizes or limited coverage of metabolic pathways. Our objective is to identify metabolites associated with RA and its cofounders using a new untargeted metabolomics platform. Moreover, to investigate the pathomechanism of RA by identifying correlations between RA-associated metabolites. 132 RA patients and 104 controls were analyzed for 927 metabolites. Metabolites were tested for association with RA using linear regression. OPLS-DA was used to discriminate RA patients from controls. Gaussian Graphical Models (GGMs) were used to identify correlated metabolites. 32 metabolites are identified as significantly (Bonferroni) associated with RA, including the previously reported metabolites as DHEAS, cortisol and androstenedione and extending that to a larger set of metabolites in the steroid pathway. RA classification using metabolic profiles shows a sensitivity of 91% and specificity of 88%. Steroid levels show variation among the RA patients according to the corticosteroid treatment; lowest in those taking the treatment at the time of the study, higher in those who never took the treatment, and highest in those who took it in the past. Finally, the GGM reflects metabolite relations from the steroidogenesis pathway.
Collapse
|
71
|
Zhang Q, Zhu B, Li Y. Resolution of Cancer-Promoting Inflammation: A New Approach for Anticancer Therapy. Front Immunol 2017; 8:71. [PMID: 28210259 PMCID: PMC5288347 DOI: 10.3389/fimmu.2017.00071] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/17/2017] [Indexed: 12/31/2022] Open
Abstract
Inflammation is a protective response that eliminates harmful stimuli and restores tissue homeostasis, whereas the failure to resolve inflammation leads to the development of malignancies. Immune cells in the tumor inflammatory microenvironment endow cancer cells with their specific hallmarks, including mutations, metabolic reprograming, angiogenesis, invasion, and metastasis. Targeting the inflammatory microenvironment with anti-inflammatory drugs (e.g., aspirin) or by enhancing antitumor immunity (e.g., chimeric antigen receptor T cell therapy) has been extensively investigated and has achieved promising results in many cancers. Recently, a novel approach promoting antitumor immunity via a dual anti-inflammatory and pro-resolving strategy was proposed based on the discovery of potent, endogenous, specialized pro-resolving mediators, including lipoxins, resolvins, protectins, and maresins. In this review, we describe the updated principal cellular and molecular mechanisms of inflammation resolution and cancer immunity and discuss the pro-resolution strategy in cancer treatment and prevention.
Collapse
Affiliation(s)
- Qi Zhang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| | - Yongsheng Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University , Chongqing , China
| |
Collapse
|
72
|
Mueller KM, Hartmann K, Kaltenecker D, Vettorazzi S, Bauer M, Mauser L, Amann S, Jall S, Fischer K, Esterbauer H, Müller TD, Tschöp MH, Magnes C, Haybaeck J, Scherer T, Bordag N, Tuckermann JP, Moriggl R. Adipocyte Glucocorticoid Receptor Deficiency Attenuates Aging- and HFD-Induced Obesity and Impairs the Feeding-Fasting Transition. Diabetes 2017; 66:272-286. [PMID: 27650854 DOI: 10.2337/db16-0381] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 09/14/2016] [Indexed: 11/13/2022]
Abstract
Glucocorticoids (GCs) are important regulators of systemic energy metabolism, and aberrant GC action is linked to metabolic dysfunctions. Yet, the extent to which normal and pathophysiological energy metabolism depend on the GC receptor (GR) in adipocytes remains unclear. Here, we demonstrate that adipocyte GR deficiency in mice significantly impacts systemic metabolism in different energetic states. Plasma metabolomics and biochemical analyses revealed a marked global effect of GR deficiency on systemic metabolite abundance and, thus, substrate partitioning in fed and fasted states. This correlated with a decreased lipolytic capacity of GR-deficient adipocytes under postabsorptive and fasting conditions, resulting from impaired signal transduction from β-adrenergic receptors to adenylate cyclase. Upon prolonged fasting, the impaired lipolytic response resulted in abnormal substrate utilization and lean mass wasting. Conversely, GR deficiency attenuated aging-/diet-associated obesity, adipocyte hypertrophy, and liver steatosis. Systemic glucose tolerance was improved in obese GR-deficient mice, which was associated with increased insulin signaling in muscle and adipose tissue. We conclude that the GR in adipocytes exerts central but diverging roles in the regulation of metabolic homeostasis depending on the energetic state. The adipocyte GR is indispensable for the feeding-fasting transition but also promotes adiposity and associated metabolic disorders in fat-fed and aged mice.
Collapse
Affiliation(s)
- Kristina M Mueller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Kerstin Hartmann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | | | - Sabine Vettorazzi
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Mandy Bauer
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Lea Mauser
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Sabine Amann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Sigrid Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Katrin Fischer
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Harald Esterbauer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH) and German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Department of Medicine, Technische Universität München, Munich, Germany
| | - Christoph Magnes
- HEALTH Institute for Biomedicine and Health Sciences, JOANNEUM RESEARCH, Forschungsgesellschaft mbH, Graz, Austria
| | | | - Thomas Scherer
- Division of Endocrinology and Metabolism, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Natalie Bordag
- Center for Biomarker Research in Medicine, CBmed GmbH, Graz, Austria
| | - Jan P Tuckermann
- Institute for Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Richard Moriggl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
- Medical University of Vienna, Vienna, Austria
| |
Collapse
|
73
|
Omics for understanding synergistic action of validamycin A and Trichoderma asperellum GDFS1009 against maize sheath blight pathogen. Sci Rep 2017; 7:40140. [PMID: 28057927 PMCID: PMC5216365 DOI: 10.1038/srep40140] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022] Open
Abstract
Sheath blight, causes by Rhizoctonia spp., threaten maize yield every year throughout the world. Trichoderma could degrade Rhizoctonia solani on maize mainly via competition and hyperparasitism, whereas validamycin A could efficiently inhibit the growth of R. solani via disturbing the energy system. By contrast, validamycin A is efficient but it takes effect in a short period, while Trichoderma takes effect in a long period though time-consuming. To overcome the disadvantages, Trichoderma asperellum GDFS1009 was used together with validamycin A. In vitro tests proved that the combined pathogen-inhibiting efficiency was significantly improved. Furthermore, results based on transcriptome and metabolome showed that validamycin A had no significant effects on growth, basic metabolism and main bio-control mechanisms of T. asperellum GDFS1009. Such few impacts may be attributed to detoxification and tolerance mechanism of T. asperellum GDFS1009. In addition, T. asperellum GDFS1009 has an ability to relieve the stress caused by validaymicn A. Meanwhile, liquid chromatography-mass spectrometry (LC-MS) results showed that only minor degradation (20%) of validamycin A was caused by T. asperellum GDFS1009 during cofermentation. All results together provide solid bases for validamycin A synergy with T. asperellum GDFS1009 in their combined biocontrol application.
Collapse
|
74
|
Mueller-Hennessen M, Düngen HD, Lutz M, Trippel TD, Kreuter M, Sigl J, Müller OJ, Tahirovic E, Witt H, Ternes P, Carvalho S, Peter E, Rein D, Schatz P, Herth F, Giannitsis E, Weis T, Frey N, Katus HA. A Novel Lipid Biomarker Panel for the Detection of Heart Failure with Reduced Ejection Fraction. Clin Chem 2016; 63:267-277. [PMID: 28062623 DOI: 10.1373/clinchem.2016.257279] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/27/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVES In this study we aimed to identify novel metabolomic biomarkers suitable for improved diagnosis of heart failure with reduced ejection fraction (HFrEF). METHODS We prospectively recruited 887 individuals consisting of HFrEF patients with either ischemic (ICMP, n = 257) or nonischemic cardiomyopathy (NICMP, n = 269), healthy controls (n = 327), and patients with pulmonary diseases (n = 34). A single-center identification (n = 238) was followed by a multicenter confirmation study (n = 649). Plasma samples from the single-center study were subjected to metabolite profiling analysis to identify metabolomic features with potential as HFrEF biomarkers. A dedicated analytical protocol was developed for the routine analysis of selected metabolic features in the multicenter cohort. RESULTS In the single-center study, 92 of 181 metabolomic features with known chemical identity (51%) were significantly changed in HFrEF patients compared to healthy controls (P <0.05). Three specific metabolomic features belonging to the lipid classes of sphingomyelins, triglycerides, and phosphatidylcholines were selected as the cardiac lipid panel (CLP) and analyzed in the multicenter study using the dedicated analytical protocol. The combination of the CLP with N-terminal pro-B-type natriuretic peptide (NT-proBNP) distinguished HFrEF patients from healthy controls with an area under the curve (AUC) of 0.97 (sensitivity 80.2%, specificity 97.6%) and was significantly superior compared to NT-proBNP alone (AUC = 0.93, sensitivity 81.7%, specificity 88.1%, P <0.001), even in the subgroups with mildly reduced left ventricular EF (0.94 vs 0.87; P <0.001) and asymptomatic patients (0.95 vs 0.91; P <0.05). CONCLUSIONS The new metabolomic biomarker panel has the potential to improve HFrEF detection, even in mild and asymptomatic stages. The observed changes further indicate lipid alterations in the setting of HFrEF.
Collapse
Affiliation(s)
- Matthias Mueller-Hennessen
- Department of Internal Medicine III, Cardiology, Angiology & Pulmonology, Heidelberg University Hospital, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Hans-Dirk Düngen
- Department of Cardiology, Charité, Campus Virchow-Klinikum, Berlin, Germany.,DZHK, Berlin, Germany
| | - Matthias Lutz
- Department of Cardiology and Angiology, University Hospital of Schleswig-Holstein, Kiel, Germany.,DZHK, Hamburg/Kiel/Lübeck, Germany
| | - Tobias Daniel Trippel
- Department of Cardiology, Charité, Campus Virchow-Klinikum, Berlin, Germany.,DZHK, Berlin, Germany
| | - Michael Kreuter
- Department of Pneumology and Respiratory Critical Care Medicine, Thoraxklinik, University of Heidelberg, and Translational Lung Research Center, Member of the German Center for Lung Research, Heidelberg, Germany
| | - Johanna Sigl
- Department of Internal Medicine III, Cardiology, Angiology & Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, Cardiology, Angiology & Pulmonology, Heidelberg University Hospital, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Elvis Tahirovic
- Department of Cardiology, Charité, Campus Virchow-Klinikum, Berlin, Germany.,DZHK, Berlin, Germany
| | | | | | | | | | | | | | - Felix Herth
- Department of Pneumology and Respiratory Critical Care Medicine, Thoraxklinik, University of Heidelberg, and Translational Lung Research Center, Member of the German Center for Lung Research, Heidelberg, Germany
| | - Evangelos Giannitsis
- Department of Internal Medicine III, Cardiology, Angiology & Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tanja Weis
- Department of Internal Medicine III, Cardiology, Angiology & Pulmonology, Heidelberg University Hospital, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| | - Norbert Frey
- Department of Cardiology and Angiology, University Hospital of Schleswig-Holstein, Kiel, Germany.,DZHK, Hamburg/Kiel/Lübeck, Germany
| | - Hugo A Katus
- Department of Internal Medicine III, Cardiology, Angiology & Pulmonology, Heidelberg University Hospital, Heidelberg, Germany; .,DZHK (German Centre for Cardiovascular Research), Heidelberg/Mannheim, Germany
| |
Collapse
|
75
|
Brown SA. Circadian Metabolism: From Mechanisms to Metabolomics and Medicine. Trends Endocrinol Metab 2016; 27:415-426. [PMID: 27113082 DOI: 10.1016/j.tem.2016.03.015] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 03/22/2016] [Accepted: 03/25/2016] [Indexed: 12/28/2022]
Abstract
The circadian clock directs nearly all aspects of diurnal physiology, including metabolism. Current research identifies several major axes by which it exerts these effects, including systemic signals as well as direct control of cellular processes by local clocks. This redundant network can transmit metabolic and timing information bidirectionally for optimal synchrony of metabolic processes. Recent advances in cellular profiling and metabolomics technologies have yielded unprecedented insights into the mechanisms behind this control. They have also helped to illuminate individual variation in these mechanisms that could prove important in personalized therapy for metabolic disease. Finally, these technologies have provided platforms with which to screen for the first potential drugs affecting clock-modulated metabolic function.
Collapse
Affiliation(s)
- Steven A Brown
- Chronobiology and Sleep Research Group, Institute of Pharmacology and Toxicology, University of Zürich, 190 Winterthurerstrasse, 8057 Zürich, Switzerland.
| |
Collapse
|
76
|
Abstract
The circadian system temporally coordinates daily rhythms in feeding behaviour and energy metabolism. The objective of the present paper is to review the mechanisms that underlie circadian regulation of lipid metabolic pathways. Circadian rhythms in behaviour and physiology are generated by master clock neurons in the suprachiasmatic nucleus (SCN). The SCN and its efferent targets in the hypothalamus integrate light and feeding signals to entrain behavioural rhythms as well as clock cells located in peripheral tissues, including the liver, adipose tissue and muscle. Circadian rhythms in gene expression are regulated at the cellular level by a molecular clock comprising a core set of clock genes/proteins. In peripheral tissues, hundreds of genes involved in lipid biosynthesis and fatty acid oxidation are rhythmically activated and repressed by clock proteins, hence providing a direct mechanism for circadian regulation of lipids. Disruption of clock gene function results in abnormal metabolic phenotypes and impaired lipid absorption, demonstrating that the circadian system is essential for normal energy metabolism. The composition and timing of meals influence diurnal regulation of metabolic pathways, with food intake during the usual rest phase associated with dysregulation of lipid metabolism. Recent studies using metabolomics and lipidomics platforms have shown that hundreds of lipid species are circadian-regulated in human plasma, including but not limited to fatty acids, TAG, glycerophospholipids, sterol lipids and sphingolipids. In future work, these lipid profiling approaches can be used to understand better the interaction between diet, mealtimes and circadian rhythms on lipid metabolism and risk for obesity and metabolic diseases.
Collapse
|
77
|
Yan B, Huang J, Zhang C, Hu X, Gao M, Shi A, Zha W, Shi L, Huang C, Yang L. Serum metabolomic profiling in patients with systemic lupus erythematosus by GC/MS. Mod Rheumatol 2016; 26:914-922. [PMID: 26915395 DOI: 10.3109/14397595.2016.1158895] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Bei Yan
- Department of Clinical Pharmacology & Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing Hospital, Beijing, P.R. China
| | - Jia Huang
- Department of Rheumatology and Immunology, Beijing Hospital, Beijing, P.R. China, and
| | - Chunmei Zhang
- Department of Rheumatology and Immunology, Beijing Hospital, Beijing, P.R. China, and
| | - Xin Hu
- Department of Clinical Pharmacology & Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing Hospital, Beijing, P.R. China
| | - Ming Gao
- Department of Rheumatology and Immunology, Beijing Hospital, Beijing, P.R. China, and
| | - Aixin Shi
- Department of Clinical Pharmacology & Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing Hospital, Beijing, P.R. China
| | - Weibin Zha
- Department of Pharmaceutics, University of Washington, Seattle, WA, USA
| | - Luyi Shi
- Department of Clinical Pharmacology & Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing Hospital, Beijing, P.R. China
| | - Cibo Huang
- Department of Rheumatology and Immunology, Beijing Hospital, Beijing, P.R. China, and
| | - Liping Yang
- Department of Clinical Pharmacology & Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, Beijing Hospital, Beijing, P.R. China
| |
Collapse
|