1
|
Yan Z, Luo J, Wang Y, Yang J, Su M, Jiang L, Yang J, Dai M, Liu A. PPARα suppresses low-intensity-noise-induced body weight gain in mice: the activated HPA axis plays an critical role. Int J Obes (Lond) 2024; 48:1274-1282. [PMID: 38902386 DOI: 10.1038/s41366-024-01550-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 05/16/2024] [Accepted: 05/21/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND As the second most risky environmental pollution, noise imposes threats to human health. Exposure to high-intensity noise causes hearing impairment, psychotic disorders, endocrine modifications. The relationship among low-intensity noise, obesity and lipid-regulating nuclear factor PPARα is not yet clear. METHODS In this study, male wild-type (WT) and Pparα-null (KO) mice on a high-fat diet (HFD) were exposed to 75 dB noise for 12 weeks to explore the effect of low-intensity noise on obesity development and the role of PPARα. 3T3-L1 cells were treated with dexamethasone (DEX) and sodium oleate (OA) to verify the down-stream effect of hypothalamic-pituitary-adrenal (HPA) axis activation on the adipose tissues. RESULTS The average body weight gain (BWG) of WT mice on HFD exposed to noise was inhibited, which was not observed in KO mice. The mass and adipocyte size of adipose tissues accounted for the above difference of BWG tendency. In WT mice on HFD, the adrenocorticotropic hormone level was increased by the noise challenge. The aggravation of fatty liver by noise exposure occurred in both mouse lines, and the transport of hepatic redundant lipid to adipose tissues were similar. The lipid metabolism in adipose tissue driven by HPA axis accorded with the BWG inhibition in vivo, validated in 3T3-L1 adipogenic stem cells. CONCLUSION Chronic exposure to low-intensity noise aggravated fatty liver in both WT and KO mice. BWG inhibition was observed only in WT mice, which covered up the aggravation of fatty liver by noise exposure. PPARα mediates the activation of HPA axis by noise exposure in mice on HFD. Elevated adrenocorticotropic hormone (ACTH) promoted lipid metabolism in adipocytes, which contributed to the disassociation of BWG and fatty liver development in male WT mice. Summary of PPARα suppresses noise-induced body weight gain in mice on high-fat-diet. Chronic exposure to low-intensity noise exposure inhibited BWG by PPARα-dependent activation of the HPA axis.
Collapse
Affiliation(s)
- Zheng Yan
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jia Luo
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Ying Wang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Jie Yang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Mingli Su
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Lei Jiang
- Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Julin Yang
- Department of Basic Nutrition, Ningbo College of Health Sciences, Ningbo, 315211, China
| | - Manyun Dai
- Health Science Center, Ningbo University, Ningbo, 315211, China.
| | - Aiming Liu
- Health Science Center, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
2
|
Linden MA, Burke SJ, Pirzadah HA, Huang TY, Batdorf HM, Mohammed WK, Jones KA, Ghosh S, Campagna SR, Collier JJ, Noland RC. Pharmacological inhibition of lipolysis prevents adverse metabolic outcomes during glucocorticoid administration. Mol Metab 2023; 74:101751. [PMID: 37295745 PMCID: PMC10300254 DOI: 10.1016/j.molmet.2023.101751] [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: 03/08/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023] Open
Abstract
OBJECTIVE Glucocorticoids are one of the most commonly prescribed classes of anti-inflammatory drugs; however, chronic treatment promotes iatrogenic (drug-induced) diabetes. As part of their physiological role, glucocorticoids stimulate lipolysis to spare glucose. We hypothesized that persistent stimulation of lipolysis during glucocorticoid therapy plays a causative role in the development of iatrogenic diabetes. METHODS Male C57BL/6J mice were given 100 μg/mL corticosterone (Cort) in the drinking water for two weeks and were fed either normal chow (TekLad 8640) or the same diet supplemented with an adipose triglyceride lipase inhibitor (Atglistatin - 2 g/kg diet) to inhibit the first step of lipolysis. RESULTS Herein, we report for the first time that glucocorticoid administration promotes a unique state of substrate excess and energetic overload in skeletal muscle that primarily results from the rampant mobilization of endogenous fuels. Inhibiting lipolysis protected mice from Cort-induced gains in fat mass, excess ectopic lipid accrual, hyperinsulinemia, and hyperglycemia. The role lipolysis plays in Cort-mediated pathology appears to differ between tissues. Within skeletal muscle, Cort-induced lipolysis facilitated diversion of glucose-derived carbons toward the pentose phosphate and hexosamine biosynthesis pathways but contributed to <3% of the Cort-induced genomic adaptations. In contrast, Cort stimulation of lipolysis accounted for ∼35% of the genomic changes in the liver but had minimal impact on hepatic metabolites reported. CONCLUSIONS These data support the idea that activation of lipolysis plays a causal role in the progression toward iatrogenic diabetes during glucocorticoid therapy with differential impact on skeletal muscle and liver.
Collapse
Affiliation(s)
- Melissa A Linden
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Department of Exercise and Health Sciences, University of Massachusetts-Boston, Boston, MA, 02125, USA.
| | - Susan J Burke
- Laboratory of Immunogenetics, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Humza A Pirzadah
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Tai-Yu Huang
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Heidi M Batdorf
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Walid K Mohammed
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Katarina A Jones
- Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN, 37916, USA.
| | - Sujoy Ghosh
- Laboratory of Computational Biology, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA; Program in Cardiovascular and Metabolic Disorders and Center for Computational Biology, Duke-National University of Singapore Medical School, Singapore, 169857, Singapore.
| | - Shawn R Campagna
- Biological and Small Molecule Mass Spectrometry Core, University of Tennessee, Knoxville, TN, 37916, USA.
| | - J Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Robert C Noland
- Skeletal Muscle Metabolism Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
3
|
Delangre E, Pommier G, Tolu S, Uzan B, Bailbé D, Movassat J. Lithium treatment mitigates the diabetogenic effects of chronic cortico-therapy. Biomed Pharmacother 2023; 164:114895. [PMID: 37224758 DOI: 10.1016/j.biopha.2023.114895] [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: 03/23/2023] [Revised: 05/09/2023] [Accepted: 05/13/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND AND PURPOSE Glucocorticoids (GCs) are the main treatment for autoimmune and inflammatory disorders and are also used as immunosuppressive therapy for patients with organ transplantation. However, these treatments have several side effects, including metabolic disorders. Indeed, cortico-therapy may induce insulin resistance, glucose intolerance, disrupted insulin and glucagon secretion, excessive gluconeogenesis, leading to diabetes in susceptible individuals. Recently, lithium has been shown to alleviate deleterious effects of GCs in various diseased conditions. EXPERIMENTAL APPROACH In this study, using two rat models of GC-induced metabolic disorders, we investigated the effects of Lithium Chloride (LiCl) in the mitigation of deleterious effects of GCs. Rats were treated either with corticosterone or dexamethasone, and with or without LiCl. Animals were then assessed for glucose tolerance, insulin sensitivity, in vivo and ex vivo glucose-induced insulin secretion and hepatic gluconeogenesis. KEY RESULTS We showed that in rats chronically treated with corticosterone, lithium treatment markedly reduced insulin resistance. In addition, in rats treated with dexamethasone, lithium administration improved glucose tolerance, associated with enhanced insulin secretion in vivo. Moreover, liver gluconeogenesis was reduced upon LiCl treatment. The improvement of insulin secretion in vivo appeared to be due to an indirect regulation of β cell function, since the ex vivo assessment of insulin secretion and β cell mass in islets from animals treated with LiCl revealed no difference compared to untreated animals. CONCLUSION AND IMPLICATIONS Collectively, our data provide evidences for the beneficial effects of lithium to mitigate the adverse metabolic effects of chronic cortico-therapy.
Collapse
Affiliation(s)
- Etienne Delangre
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Gaëlle Pommier
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France; Université Paris Cité, UFR Sciences du Vivant, F-75013 Paris, France
| | - Stefania Tolu
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Benjamin Uzan
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Danielle Bailbé
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France
| | - Jamileh Movassat
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, F-75013 Paris, France.
| |
Collapse
|
4
|
Antevska A, Long CC, Dupuy SD, Collier JJ, Karlstad MD, Do TD. Mouse Pancreatic Peptide Hormones Probed at the Sub-Single-Islet Level: The Effects of Acute Corticosterone Treatment. J Proteome Res 2023; 22:235-245. [PMID: 36412564 DOI: 10.1021/acs.jproteome.2c00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
We combine liquid chromatography coupled with ion mobility spectrometry-mass spectrometry to elucidate how short exposure to corticosterone (Cort) alters the output of mouse pancreatic islet hormones. The workflow enables the robust separation of mouse insulin 1 (Ins1) and insulin 2 (Ins2) and the detection of major islet hormones in a homogenate equivalent to 100-150 islet cells. We show that Ins2 has a unique structure and is degraded much faster than Ins1. Further investigation indicates that Ins2 may populate both T and R states, whereas Ins1 may not. The assemblies of Ins1's B-chain also introduce more structural heterogeneity than Ins2. Collectively, these features account for their unique degradation profiles, the diabetes risk associated with Ins1, and the protective effect of Ins2. In the same experiments, we observe that the ratio of amylin to Ins1 increased significantly in Cort-treated mice (15:1) compared to the control mice (42:1), correlating well with β-cell proliferation observed in immunoassays on the same animal model. We observe no increase in intact full-length insulin levels but more of the truncated forms, indicating that enzymatic activity is accelerated. Our data provide a molecular basis for reduced insulin action induced by Cort and connections between insulin turnover and insulin resistance.
Collapse
Affiliation(s)
- Aleksandra Antevska
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee37996, United States
| | - Connor C Long
- Department of Biochemistry, Cellular, and Molecular Biology, University of Tennessee, Knoxville, Tennessee37996, United States
| | - Samuel D Dupuy
- Department of Surgery, Graduate School of Medicine, University of Tennessee, Knoxville, Tennessee37996, United States
| | - J Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, Louisiana70808, United States
| | - Michael D Karlstad
- Department of Surgery, Graduate School of Medicine, University of Tennessee, Knoxville, Tennessee37996, United States
| | - Thanh D Do
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee37996, United States
| |
Collapse
|
5
|
Helman TJ, Headrick JP, Stapelberg NJC, Braidy N. The sex-dependent response to psychosocial stress and ischaemic heart disease. Front Cardiovasc Med 2023; 10:1072042. [PMID: 37153459 PMCID: PMC10160413 DOI: 10.3389/fcvm.2023.1072042] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 04/03/2023] [Indexed: 05/09/2023] Open
Abstract
Stress is an important risk factor for modern chronic diseases, with distinct influences in males and females. The sex specificity of the mammalian stress response contributes to the sex-dependent development and impacts of coronary artery disease (CAD). Compared to men, women appear to have greater susceptibility to chronic forms of psychosocial stress, extending beyond an increased incidence of mood disorders to include a 2- to 4-fold higher risk of stress-dependent myocardial infarction in women, and up to 10-fold higher risk of Takotsubo syndrome-a stress-dependent coronary-myocardial disorder most prevalent in post-menopausal women. Sex differences arise at all levels of the stress response: from initial perception of stress to behavioural, cognitive, and affective responses and longer-term disease outcomes. These fundamental differences involve interactions between chromosomal and gonadal determinants, (mal)adaptive epigenetic modulation across the lifespan (particularly in early life), and the extrinsic influences of socio-cultural, economic, and environmental factors. Pre-clinical investigations of biological mechanisms support distinct early life programming and a heightened corticolimbic-noradrenaline-neuroinflammatory reactivity in females vs. males, among implicated determinants of the chronic stress response. Unravelling the intrinsic molecular, cellular and systems biological basis of these differences, and their interactions with external lifestyle/socio-cultural determinants, can guide preventative and therapeutic strategies to better target coronary heart disease in a tailored sex-specific manner.
Collapse
Affiliation(s)
- Tessa J. Helman
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
- Correspondence: Tessa J. Helman
| | - John P. Headrick
- Schoolof Pharmacy and Medical Sciences, Griffith University, Southport, QLD, Australia
| | | | - Nady Braidy
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, NSW, Sydney, Australia
| |
Collapse
|
6
|
Zhang Y, Shen T, Wang S. Progression from prediabetes to type 2 diabetes mellitus induced by overnutrition. Hormones (Athens) 2022; 21:591-597. [PMID: 36197636 DOI: 10.1007/s42000-022-00399-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/19/2022] [Indexed: 12/31/2022]
Abstract
Prediabetes has developed into a global pandemic, its prevalence increasing year by year. Although lifestyle changes are advocated as the basis for prediabetes treatment, some patients fail to choose or adhere to appropriate interventions. The basis for selecting an appropriate intervention is determining the stage and cause of the disease. In this review, we aimed to examine the various types and disease processes of prediabetes caused by overnutrition, the present review supporting the hypothesis that overnutrition-induced hyperinsulinemia precedes insulin resistance (IR) and independently causes β-cell dysfunction. Tissue insulin resistance is the main feature of prediabetes with the crosstalk between tissues promoting the formation of systemic insulin resistance. Finally, both β-cell dysfunction induced by hyperinsulinemia or IR and reduced β-cell mass can lead to abnormal insulin secretion and contribute to development of type 2 diabetes mellitus (T2DM). Hence, overnutrition can cause multiple prediabetes phenotypes resulting in development of T2DM through different trajectories. Future diagnosis and treatment should therefore more carefully consider the disease phenotype and stage of development in patients with prediabetes to reduce the incidence of T2DM.
Collapse
Affiliation(s)
- Yuli Zhang
- School of Physical Education & Sports Science, South China Normal University, No.55, West of Zhongshan Ave., Tianhe District, Guangzhou City, 510006, Guangdong Province, China
| | - Tuming Shen
- School of Physical Education & Sports Science, South China Normal University, No.55, West of Zhongshan Ave., Tianhe District, Guangzhou City, 510006, Guangdong Province, China
| | - Songtao Wang
- School of Physical Education & Sports Science, South China Normal University, No.55, West of Zhongshan Ave., Tianhe District, Guangzhou City, 510006, Guangdong Province, China.
| |
Collapse
|
7
|
Maternal stress induced endoplasmic reticulum stress and impaired pancreatic islets’ insulin secretion via glucocorticoid receptor upregulation in adult male rat offspring. Sci Rep 2022; 12:12552. [PMID: 35869151 PMCID: PMC9307850 DOI: 10.1038/s41598-022-16621-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 07/12/2022] [Indexed: 11/23/2022] Open
Abstract
Exposure to perinatal (prenatal and/or postnatal) stress is considered as a risk factor for metabolic disorders in later life. Accordingly, this study aimed to investigate the perinatal stress effects on the pancreatic endoplasmic reticulum (ER) stress induction, insulin secretion impairment and WFS1 (wolframin ER transmembrane Glycoprotein, which is involved in ER homeostasis and insulin secretion) expression changes, in rat offspring. According to the dams’ period of exposure to variable stress, their male offspring were divided into, control (CTRL); pre-pregnancy, pregnancy, lactation stress (PPPLS); pre-pregnancy stress (PPS); pregnancy stress (PS); lactation stress (LS); pre-pregnancy, pregnancy stress (PPPS); pregnancy, lactation stress (PLS); pre-pregnancy, lactation stress (PPLS) groups. Offspring pancreases were removed for ER extraction and the assessment of ER stress biomarkers, WFS1 gene DNA methylation, and isolated islets’ insulin secretion. Glucose tolerance was also tested. In the stressed groups, maternal stress significantly increased plasma corticosterone levels. In PPS, PS, and PPPS groups, maternal stress increased Bip (Hsp70; heat shock protein family A member 4), Chop (Ddit3; DNA- damage inducible transcript3), and WFS1 protein levels in pancreatic extracted ER. Moreover, the islets’ insulin secretion and content along with glucose tolerance were impaired in these groups. In PPS, PS, LS and PPPS groups, the pancreatic glucocorticoid receptor (GR) expression increased. Maternal stress did not affect pancreatic WFS1 DNA methylation. Thus, maternal stress, during prenatal period, impaired the islets’ insulin secretion and glucose homeostasis in adult male offspring, possibly through the induction of ER stress and GR expression in the pancreas, in this regard the role of WFS1 protein alteration in pancreatic ER should also be considered.
Collapse
|
8
|
Bel JS, Tai TC, Khaper N, Lees SJ. Chronic glucocorticoid exposure causes brown adipose tissue whitening, alters whole-body glucose metabolism and increases tissue uncoupling protein-1. Physiol Rep 2022; 10:e15292. [PMID: 35510321 PMCID: PMC9069169 DOI: 10.14814/phy2.15292] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/05/2022] [Accepted: 04/11/2022] [Indexed: 04/14/2023] Open
Abstract
Adipose tissue (AT) has been found to exist in two predominant forms, white and brown. White adipose tissue (WAT) is the body's conventional storage organ, and brown adipose tissue (BAT) is responsible for non-shivering thermogenesis which allows mammals to produce heat and regulate body temperature. Studies examining BAT and its role in whole-body metabolism have found that active BAT utilizes glucose and circulating fatty acids and is associated with improved metabolic outcomes. While the beiging of WAT is a growing area of interest, the possibility of the BAT depot to "whiten" and store more triglycerides also has metabolic and health implications. Currently, there are limited studies that examine the effects of chronic stress and its ability to induce a white-like phenotype in the BAT depot. This research examined how chronic exposure to the murine stress hormone, corticosterone, for 4 weeks can affect the whitening process of BAT in C57BL/6 male mice. Separate treatments with mirabegron, a known β3-adrenergic receptor agonist, were used to directly compare the effects of corticosterone with a beiging phenotype. Corticosterone-treated mice had significantly higher body weight (p ≤ 0.05) and BAT mass (p ≤ 0.05), increased adipocyte area (p ≤ 0.05), were insulin resistant (p ≤ 0.05), and significantly elevated expressions of uncoupling protein 1 (UCP-1) in BAT (p ≤ 0.05) while mitochondrial content remained unchanged. This whitened phenotype has not been previously associated with increased uncoupling proteins under chronic stress and may represent a compensatory mechanism being initiated under these conditions. These findings have implications for the study of BAT in response to chronic glucocorticoid exposure potentially leading to BAT dysfunction and negative impacts on whole-body glucose metabolism.
Collapse
Affiliation(s)
- Jocelyn S. Bel
- Biotechnology ProgramLakehead UniversityThunder BayOntarioCanada
| | - T. C. Tai
- Northern Ontario School of MedicineThunder BayOntarioCanada
- BiologyLaurentian UniversitySudburyOntarioCanada
- Chemistry and BiochemistryLaurentian UniversitySudburyOntarioCanada
- Biomolecular Sciences ProgramLaurentian UniversitySudburyOntarioCanada
| | - Neelam Khaper
- Northern Ontario School of MedicineThunder BayOntarioCanada
- Biomolecular Sciences ProgramLaurentian UniversitySudburyOntarioCanada
- BiologyLakehead UniversityThunder BayOntarioCanada
| | - Simon J. Lees
- Northern Ontario School of MedicineThunder BayOntarioCanada
- BiologyLakehead UniversityThunder BayOntarioCanada
| |
Collapse
|
9
|
Relationship between Insulin Secretion and Arterial Stiffness in Essential Hypertension. Int J Hypertens 2022; 2021:5015797. [PMID: 34976408 PMCID: PMC8720019 DOI: 10.1155/2021/5015797] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/08/2021] [Indexed: 01/17/2023] Open
Abstract
The study aims to explore the relationship between plasma insulin secretion and arterial stiffness in nondiabetic essential hypertensive patients. A total of 730 nondiabetic essential hypertensive patients registered between January 2016 and October 2020 were enrolled. A two-hour oral glucose tolerance test (OGTT) was performed to detect the levels of C-peptide and blood glucose at 0 hours and 2 hours, as well as the difference between C-peptide (Δ C-peptide) and blood glucose (Δ blood glucose) over the same period. Patients were divided into two groups: the normal glucose tolerance (NGT) group (n = 322) and the impaired glucose tolerance (IGT) group (n = 408). A multiple linear regression analysis was used to evaluate the association between brachial-ankle pulse wave velocity (baPWV) and the other factors. 0 h C-peptide, 2 h C-peptide, and Δ C-peptide were found to be higher in the IGT group. baPWV was positively linear correlated with 2 h C-peptide (r = 0.086, p=0.020) and Δ C-peptide (r = 0.115, p=0.002). baPWV remained independently associated with 0 h C-peptide, 2 h C-peptide, and Δ C-peptide, after adjusting by age, gender, smoking, body mass index (BMI), high-density lipoprotein (HDL), cholesterol, systolic blood pressure (SBP), and triglycerides (TG). Our data shows that higher endogenous insulin secretion might play an important role in the progression of arterial stiffness in nondiabetic essential hypertensive patients.
Collapse
|
10
|
Maudsley S, Leysen H, van Gastel J, Martin B. Systems Pharmacology: Enabling Multidimensional Therapeutics. COMPREHENSIVE PHARMACOLOGY 2022:725-769. [DOI: 10.1016/b978-0-12-820472-6.00017-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
11
|
Leysen H, Walter D, Christiaenssen B, Vandoren R, Harputluoğlu İ, Van Loon N, Maudsley S. GPCRs Are Optimal Regulators of Complex Biological Systems and Orchestrate the Interface between Health and Disease. Int J Mol Sci 2021; 22:ijms222413387. [PMID: 34948182 PMCID: PMC8708147 DOI: 10.3390/ijms222413387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 02/06/2023] Open
Abstract
GPCRs arguably represent the most effective current therapeutic targets for a plethora of diseases. GPCRs also possess a pivotal role in the regulation of the physiological balance between healthy and pathological conditions; thus, their importance in systems biology cannot be underestimated. The molecular diversity of GPCR signaling systems is likely to be closely associated with disease-associated changes in organismal tissue complexity and compartmentalization, thus enabling a nuanced GPCR-based capacity to interdict multiple disease pathomechanisms at a systemic level. GPCRs have been long considered as controllers of communication between tissues and cells. This communication involves the ligand-mediated control of cell surface receptors that then direct their stimuli to impact cell physiology. Given the tremendous success of GPCRs as therapeutic targets, considerable focus has been placed on the ability of these therapeutics to modulate diseases by acting at cell surface receptors. In the past decade, however, attention has focused upon how stable multiprotein GPCR superstructures, termed receptorsomes, both at the cell surface membrane and in the intracellular domain dictate and condition long-term GPCR activities associated with the regulation of protein expression patterns, cellular stress responses and DNA integrity management. The ability of these receptorsomes (often in the absence of typical cell surface ligands) to control complex cellular activities implicates them as key controllers of the functional balance between health and disease. A greater understanding of this function of GPCRs is likely to significantly augment our ability to further employ these proteins in a multitude of diseases.
Collapse
Affiliation(s)
- Hanne Leysen
- Receptor Biology Lab, University of Antwerp, 2610 Wilrijk, Belgium; (H.L.); (D.W.); (B.C.); (R.V.); (İ.H.); (N.V.L.)
| | - Deborah Walter
- Receptor Biology Lab, University of Antwerp, 2610 Wilrijk, Belgium; (H.L.); (D.W.); (B.C.); (R.V.); (İ.H.); (N.V.L.)
| | - Bregje Christiaenssen
- Receptor Biology Lab, University of Antwerp, 2610 Wilrijk, Belgium; (H.L.); (D.W.); (B.C.); (R.V.); (İ.H.); (N.V.L.)
| | - Romi Vandoren
- Receptor Biology Lab, University of Antwerp, 2610 Wilrijk, Belgium; (H.L.); (D.W.); (B.C.); (R.V.); (İ.H.); (N.V.L.)
| | - İrem Harputluoğlu
- Receptor Biology Lab, University of Antwerp, 2610 Wilrijk, Belgium; (H.L.); (D.W.); (B.C.); (R.V.); (İ.H.); (N.V.L.)
- Department of Chemistry, Middle East Technical University, Çankaya, Ankara 06800, Turkey
| | - Nore Van Loon
- Receptor Biology Lab, University of Antwerp, 2610 Wilrijk, Belgium; (H.L.); (D.W.); (B.C.); (R.V.); (İ.H.); (N.V.L.)
| | - Stuart Maudsley
- Receptor Biology Lab, University of Antwerp, 2610 Wilrijk, Belgium; (H.L.); (D.W.); (B.C.); (R.V.); (İ.H.); (N.V.L.)
- Correspondence:
| |
Collapse
|
12
|
Pioglitazone Reverses Markers of Islet Beta-Cell De-Differentiation in db/db Mice While Modulating Expression of Genes Controlling Inflammation and Browning in White Adipose Tissue from Insulin-Resistant Mice and Humans. Biomedicines 2021; 9:biomedicines9091189. [PMID: 34572374 PMCID: PMC8470788 DOI: 10.3390/biomedicines9091189] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/21/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity, insulin resistance, and type 2 diabetes contribute to increased morbidity and mortality in humans. The db/db mouse is an important mouse model that displays many key features of the human disease. Herein, we used the drug pioglitazone, a thiazolidinedione with insulin-sensitizing properties, to investigate blood glucose levels, indicators of islet β-cell health and maturity, and gene expression in adipose tissue. Oral administration of pioglitazone lowered blood glucose levels in db/db mice with a corresponding increase in respiratory quotient, which indicates improved whole-body carbohydrate utilization. In addition, white adipose tissue from db/db mice and from humans treated with pioglitazone showed increased expression of glycerol kinase. Both db/db mice and humans given pioglitazone displayed increased expression of UCP-1, a marker typically associated with brown adipose tissue. Moreover, pancreatic β-cells from db/db mice treated with pioglitazone had greater expression of insulin and Nkx6.1 as well as reduced abundance of the de-differentiation marker Aldh1a3. Collectively, these findings indicate that four weeks of pioglitazone therapy improved overall metabolic health in db/db mice. Our data are consistent with published reports of human subjects administered pioglitazone and with analysis of human adipose tissue taken from subjects treated with pioglitazone. In conclusion, the current study provides evidence that pioglitazone restores key markers of metabolic health and also showcases the utility of the db/db mouse to understand mechanisms associated with human metabolic disease and interventions that provide therapeutic benefit.
Collapse
|
13
|
Aravani D, Kassi E, Chatzigeorgiou A, Vakrou S. Cardiometabolic Syndrome: An Update on Available Mouse Models. Thromb Haemost 2021; 121:703-715. [PMID: 33280078 DOI: 10.1055/s-0040-1721388] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiometabolic syndrome (CMS), a disease entity characterized by abdominal obesity, insulin resistance (IR), hypertension, and hyperlipidemia, is a global epidemic with approximately 25% prevalence in adults globally. CMS is associated with increased risk for cardiovascular disease (CVD) and development of diabetes. Due to its multifactorial etiology, the development of several animal models to simulate CMS has contributed significantly to the elucidation of the disease pathophysiology and the design of therapies. In this review we aimed to present the most common mouse models used in the research of CMS. We found that CMS can be induced either by genetic manipulation, leading to dyslipidemia, lipodystrophy, obesity and IR, or obesity and hypertension, or by administration of specific diets and drugs. In the last decade, the ob/ob and db/db mice were the most common obesity and IR models, whereas Ldlr-/- and Apoe-/- were widely used to induce hyperlipidemia. These mice have been used either as a single transgenic or combined with a different background with or without diet treatment. High-fat diet with modifications is the preferred protocol, generally leading to increased body weight, hyperlipidemia, and IR. A plethora of genetically engineered mouse models, diets, drugs, or synthetic compounds that are available have advanced the understanding of CMS. However, each researcher should carefully select the most appropriate model and validate its consistency. It is important to consider the differences between strains of the same animal species, different animals, and most importantly differences to human when translating results.
Collapse
Affiliation(s)
- Dimitra Aravani
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Styliani Vakrou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Department of Cardiology, "Laiko" General Hospital, Athens, Greece
| |
Collapse
|
14
|
Yang HJ, Koh E, Sung MK, Kang H. Changes Induced by Mind-Body Intervention Including Epigenetic Marks and Its Effects on Diabetes. Int J Mol Sci 2021; 22:ijms22031317. [PMID: 33525677 PMCID: PMC7865217 DOI: 10.3390/ijms22031317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/23/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022] Open
Abstract
Studies have evidenced that epigenetic marks associated with type 2 diabetes (T2D) can be inherited from parents or acquired through fetal and early-life events, as well as through lifelong environments or lifestyles, which can increase the risk of diabetes in adulthood. However, epigenetic modifications are reversible, and can be altered through proper intervention, thus mitigating the risk factors of T2D. Mind-body intervention (MBI) refers to interventions like meditation, yoga, and qigong, which deal with both physical and mental well-being. MBI not only induces psychological changes, such as alleviation of depression, anxiety, and stress, but also physiological changes like parasympathetic activation, lower cortisol secretion, reduced inflammation, and aging rate delay, which are all risk factors for T2D. Notably, MBI has been reported to reduce blood glucose in patients with T2D. Herein, based on recent findings, we review the effects of MBI on diabetes and the mechanisms involved, including epigenetic modifications.
Collapse
Affiliation(s)
- Hyun-Jeong Yang
- Korea Institute of Brain Science, Seoul 06022, Korea; (M.-K.S.); (H.K.)
- Department of Integrative Health Care, University of Brain Education, Cheonan 31228, Korea
- Correspondence:
| | - Eugene Koh
- Temasek Life Sciences Laboratories, Singapore 117604, Singapore;
| | - Min-Kyu Sung
- Korea Institute of Brain Science, Seoul 06022, Korea; (M.-K.S.); (H.K.)
| | - Hojung Kang
- Korea Institute of Brain Science, Seoul 06022, Korea; (M.-K.S.); (H.K.)
| |
Collapse
|
15
|
Santos CD, da Silva JS, Brunetta HS, Chagas TR, Zoccal DB, Nunes EA, Rafacho A. Impact of combined long-term fructose and prednisolone intake on glucose and lipid homeostasis in rats: benefits of intake interruption or fish oil administration. J Nutr Biochem 2020; 90:108572. [PMID: 33388348 DOI: 10.1016/j.jnutbio.2020.108572] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/19/2020] [Accepted: 11/24/2020] [Indexed: 12/14/2022]
Abstract
We investigated whether combined long-term fructose and prednisolone intake would be more detrimental to the glucose homeostasis than if ingested separately. We also evaluated whether fish oil administration or interruption of treatments has any positive impact. For this, male adult Wistar rats ingested fructose (20%) (F) or prednisolone (12.5 µg/mL) (P) or both (FP) through drinking water for 12 weeks. A separate group of fructose and prednisolone-treated rats received fish oil treatment (1 g/kg) in the last 6 weeks. In another group, the treatment with fructose and prednisolone was interrupted after 12 weeks, and the animals were followed for more 12 weeks. Control groups ran in parallel (C). The F group had higher plasma TG (+42%) and visceral adiposity (+63%), whereas the P group had lower insulin sensitivity (-33%) and higher insulinemia (+200%). Only the the FP group developed these alterations combined with higher circulating uric acid (+126%), hepatic triacylglycerol content (+16.2-fold), lipid peroxidation (+173%) and lower catalase activity (-32%) that were associated with lower protein kinase B content and AMP-activated protein kinase (AMPK) phosphorylation in the liver, lower AMPK phosphorylation in the adipose tissue and higher beta-cell mass. Fish oil ingestion attenuated the elevation in circulating triacylglycerol and uric acid values, while the interruption of sugar and glucocorticoid intake reverted almost all modified parameters. In conclusion, long-term intake of fructose and prednisolone by male rats are more detrimental to glucose and lipid homeostasis than if ingested separately and the benefits of treatment interruption are broader than fish oil treatment.
Collapse
Affiliation(s)
- Cristiane Dos Santos
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil; Multicentre Graduate Program in Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Julia Spanhol da Silva
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil; Multicentre Graduate Program in Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Henver Simionato Brunetta
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil; Multicentre Graduate Program in Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Thays Rodrigues Chagas
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil; Graduate Program in Nutrition, Centre of Health Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Daniel Breseghello Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University - UNESP, Araraquara, São Paulo, Brazil
| | - Everson Araújo Nunes
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil; Multicentre Graduate Program in Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil; Graduate Program in Nutrition, Centre of Health Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil
| | - Alex Rafacho
- Laboratory of Investigation in Chronic Diseases - LIDoC, Department of Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil; Multicentre Graduate Program in Physiological Sciences, Centre of Biological Sciences, Federal University of Santa Catarina - UFSC, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
16
|
Fouda YB, Ngo Lemba Tom E, Atsamo AD, Bonabe C, Dimo T. Effects of stem bark aqueous extract of Fagara tessmannii Engl (Rutaceae) on cardiovascular risks related to monosodium glutamate-induced obesity in rat: In vivo and in vitro assessments. JOURNAL OF ETHNOPHARMACOLOGY 2020; 260:112972. [PMID: 32446928 DOI: 10.1016/j.jep.2020.112972] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/22/2020] [Accepted: 05/09/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fagara tessmannii is a shrub of the African rainforests in South-West, Centre, South and East provinces in Cameroon. It is used in traditional medicine for the treatment of tumors, swellings, inflammation, gonorrhoea, schistosomiasis, antifungal, heart diseases and as anti-hypertensive. AIM OF THE STUDY We investigated the potential effects of F. tessmannii on cardiovascular risk related to monosodium glutamate-induced obesity. MATERIALS AND METHODS Monosodium glutamate (MSG, 4 mg/g/day) was injected subcutaneously to newborn Wistar rats for the four consecutive first days of their life and on the 6th, 8th and 10th day after birth. After 21 weeks, obese rats were treated orally with F. tessmannii (100 or 200 mg/kg/day), orlistat (10 mg/kg/day) or telmisartan (10 mg/kg/day) for 6 weeks. Body weight, obesity, body mass index (BMI), Lee index, insulin sensitivity and glucose tolerance, blood pressure, lipid profile as a Coronary Risk Index (CRI), and reactivity of isolated thoracic aorta were evaluated. RESULTS In addition to significantly decrease body weight (17.60% and 20.34%), BMI, Lee's index, retroperitoneal fat, total adiposity, and coronary risk indicators, F. tessmannii has significantly decreased insulin resistance and hyperglycemia and high blood pressure observed in MSG-obese rats. The high contractility to phenylephrine as well as the hypersensitivity to sodium nitroprusside (a nitric oxide-donor), observed in MSG aortic rings were significantly reduced by the F. tessmannii extract. Enhanced serum Na+ and Cl- levels and decreased K+ observed in obese rats were also significantly reversed after F. tessmannii treatment. CONCLUSIONS F. tessmannii fights against obesity and associated cardiovascular risks by modulating production and vascular responsiveness to vasoactive factors, monitoring premature aging. F. tessmannii promotes the loss of ectopic fat and other fatty tissues, the sensitivity of the peripherical tissues to insulin, the energy expenditure and the renovascular decompression and regulates ions movement which prevents hypertension.
Collapse
Affiliation(s)
- Yannick Bekono Fouda
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon.
| | - Esther Ngo Lemba Tom
- Department of Biological Sciences, Higher Teachers' Training College, University of Yaoundé I, P.O. Box 47, Yaoundé, Cameroon.
| | - Albert Donatien Atsamo
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon.
| | - Christian Bonabe
- Department of Biological Sciences, Faculty of Sciences, University of Ngaoundéré, P.O. Box 454, Ngaoundéré, Cameroon.
| | - Théophile Dimo
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, University of Yaoundé I, P.O. Box 812, Yaoundé, Cameroon.
| |
Collapse
|
17
|
Miranda MA, Carson C, St. Pierre CL, Macias‐Velasco JF, Hughes JW, Kunzmann M, Schmidt H, Wayhart JP, Lawson HA. Spontaneous restoration of functional β-cell mass in obese SM/J mice. Physiol Rep 2020; 8:e14573. [PMID: 33113267 PMCID: PMC7592878 DOI: 10.14814/phy2.14573] [Citation(s) in RCA: 4] [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: 08/06/2020] [Accepted: 08/08/2020] [Indexed: 12/23/2022] Open
Abstract
Maintenance of functional β-cell mass is critical to preventing diabetes, but the physiological mechanisms that cause β-cell populations to thrive or fail in the context of obesity are unknown. High fat-fed SM/J mice spontaneously transition from hyperglycemic-obese to normoglycemic-obese with age, providing a unique opportunity to study β-cell adaptation. Here, we characterize insulin homeostasis, islet morphology, and β-cell function during SM/J's diabetic remission. As they resolve hyperglycemia, obese SM/J mice dramatically increase circulating and pancreatic insulin levels while improving insulin sensitivity. Immunostaining of pancreatic sections reveals that obese SM/J mice selectively increase β-cell mass but not α-cell mass. Obese SM/J mice do not show elevated β-cell mitotic index, but rather elevated α-cell mitotic index. Functional assessment of isolated islets reveals that obese SM/J mice increase glucose-stimulated insulin secretion, decrease basal insulin secretion, and increase islet insulin content. These results establish that β-cell mass expansion and improved β-cell function underlie the resolution of hyperglycemia, indicating that obese SM/J mice are a valuable tool for exploring how functional β-cell mass can be recovered in the context of obesity.
Collapse
Affiliation(s)
- Mario A. Miranda
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | - Caryn Carson
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | | | | | - Jing W. Hughes
- Department of MedicineWashington University School of MedicineSaint LouisMOUSA
| | - Marcus Kunzmann
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | - Heather Schmidt
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | - Jessica P. Wayhart
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| | - Heather A. Lawson
- Department of GeneticsWashington University School of MedicineSaint LouisMOUSA
| |
Collapse
|
18
|
Adeyanju OA, Michael OS, Soladoye AO, Olatunji LA. Blockade of mineralocorticoid receptor ameliorates oral contraceptive-induced insulin resistance by suppressing elevated uric acid and glycogen synthase kinase-3 instead of circulating mineralocorticoid. Arch Physiol Biochem 2020; 126:225-234. [PMID: 30318954 DOI: 10.1080/13813455.2018.1509220] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Context: Estrogen-progestin combined oral contraceptive (COC) has been connected to mineralocorticoid receptor (MR) activation and adverse cardiometabolic events. We consequently hypothesised that insulin resistance (IR), hyperuricemia, and elevated circulating GSK-3 induced by COC is through activation of MR via mineralocorticoid and glucocorticoid pathways.Methods: Female Wistar rats aged 12 weeks received (po) vehicle and COC (1.0 μg ethinylestradiol plus 5.0 μg levonorgestrel) with or without MR blocker (0.25 mg/kg spironolactone; Spl), daily for eight weeks.Results: Data showed that COC treatment led to increased IR, 1-hour postload glucose level, insulinemia, triglyceride/HDL-cholesterol ratio, total cholesterol/HDL-cholesterol ratio, uric acid, GSK-3, aldosterone, corticosterone values, impaired glucose tolerance and pancreatic β-cell function. However, MR blockade by Spl ameliorated all these alterations except that of aldosterone.Conclusion: The results demonstrate that COC induces IR, hyperuricemia and high GSK-3 levels through activation of MR via glucocorticoid dependent pathway.
Collapse
Affiliation(s)
- O A Adeyanju
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria
- Cardiometabolic Research Unit, Department of Physiology, College of Medicine and Health sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - O S Michael
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria
- Cardiometabolic Research Unit, Department of Physiology, College of Health sciences, Bowen University, Iwo, Nigeria
| | - A O Soladoye
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria
- Cardiometabolic Research Unit, Department of Physiology, College of Health sciences, Bowen University, Iwo, Nigeria
| | - L A Olatunji
- HOPE Cardiometabolic Research Team, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
19
|
Kotańska M, Mika K, Sałaciak K, Wheeler L, Sapa J, Kieć-Kononowicz K, Pytka K. Pitolisant protects mice chronically treated with corticosterone from some behavioral but not metabolic changes in corticosterone-induced depression model. Pharmacol Biochem Behav 2020; 196:172974. [PMID: 32565240 DOI: 10.1016/j.pbb.2020.172974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/09/2020] [Accepted: 06/17/2020] [Indexed: 01/12/2023]
Abstract
PURPOSE Histamine H3 receptor ligands may have antidepressant and anxiolytic effects. They can also compensate for metabolic disorders, which affect glucose or triglyceride levels. In previous studies, we have shown that pitolisant, a histamine H3 receptor antagonist/inverse agonist and σ1 receptor agonist, prevented the development of certain metabolic and depressive-like disorders in mice that have been treated chronically with olanzapine. METHODS As a continuation of our previous experiments, this study aimed to investigate the antidepressant- and anxiolytic-like activity of pitolisant in mice using the corticosterone-induced depression model. The forced swim and the elevated plus maze tests were used as behavioral endpoints. We also studied the effect pitolisant had on the level of acetoacetic acid in the urine as well as the glucose tolerance and body weight of the mice that had been administered corticosterone. RESULTS Pitolisant (10 mg/kg b.w.) did not prevent depressive-like behavior in mice during the chronic corticosterone administration but did counteract anxiety-like behavior, whilst fluoxetine (10 mg/kg) was shown to protect the mice from both of these behaviors. None of the treatments that were used in the study showed an effect on the locomotor activity of the mice. Pitolisant did not prevent an increase in acetoacetic acid levels in the urine, nor did it improve glucose tolerance in the tested mice. CONCLUSION Although literature data indicates that there is significant potential for finding an antidepressant and anti-diabetic drug among the histamine H3 and σ1 receptor ligands, in our study, pitolisant was shown to only slightly compensate for corticosterone-induced abnormalities. However, further research will be required to study pitolisant's anxiolytic-like activity.
Collapse
Affiliation(s)
- Magdalena Kotańska
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, PL 30-688 Krakow, Poland.
| | - Kamil Mika
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, PL 30-688 Krakow, Poland
| | - Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Lee Wheeler
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Jacek Sapa
- Department of Pharmacological Screening, Jagiellonian University Medical College, 9 Medyczna Street, PL 30-688 Krakow, Poland
| | - Katarzyna Kieć-Kononowicz
- Department of Technology and Biotechnology of Drugs, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
20
|
Hudish LI, Reusch JE, Sussel L. β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes. J Clin Invest 2020; 129:4001-4008. [PMID: 31424428 DOI: 10.1172/jci129188] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In a society where physical activity is limited and food supply is abundant, metabolic diseases are becoming a serious epidemic. Metabolic syndrome (MetS) represents a cluster of metabolically related symptoms such as obesity, hypertension, dyslipidemia, and carbohydrate intolerance, and significantly increases type 2 diabetes mellitus risk. Insulin resistance and hyperinsulinemia are consistent characteristics of MetS, but which of these features is the initiating insult is still widely debated. Regardless, both of these conditions trigger adverse responses from the pancreatic β cell, which is responsible for producing, storing, and releasing insulin to maintain glucose homeostasis. The observation that the degree of β cell dysfunction correlates with the severity of MetS highlights the need to better understand β cell dysfunction in the development of MetS. This Review focuses on the current understanding from rodent and human studies of the progression of β cell responses during the development of MetS, as well as recent findings addressing the complexity of β cell identity and heterogeneity within the islet during disease progression. The differential responses observed in β cells together with the heterogeneity in disease phenotypes within the patient population emphasize the need to better understand the mechanisms behind β cell adaptation, identity, and dysfunction in MetS.
Collapse
Affiliation(s)
| | - Jane Eb Reusch
- Division of Endocrinology, University of Colorado Anschutz Medical Center, Aurora, Colorado, USA
| | | |
Collapse
|
21
|
Aremu AO, Lilian DC, Olufemi SA, Aderemi OL. Combined but not single treatment with ethinylestradiol/levonorgestrel and spironolactone reduces plasminogen activator inhibitor-1 in insulin-resistant ovariectomised rats. J Renin Angiotensin Aldosterone Syst 2019; 20:1470320319895933. [PMID: 31856649 PMCID: PMC6927203 DOI: 10.1177/1470320319895933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Objective: Increased circulating level of plasminogen activator inhibitor-1 (PAI-1) is
associated with menopausal oestrogen deficiency. We therefore hypothesised
that the combined oral contraceptive (COC) with spironolactone (SPL)
improves insulin resistance (IR) in ovariectomised (OVX) rats by reducing
circulating PAI-1. Methods: Twelve-week-old female Wistar rats were divided into sham-operated (SHM),
OVX, OVX+SPL (0.25 mg/kg), COC (1.0 µg ethinylestradiol and 5.0 µg
levonorgestrel) and OVX+COC+SPL rats treated with COC and SPL daily for
eight weeks. IR was assessed by homeostatic model assessment of IR
(HOMA-IR). Results: Data showed that OVX rats had a higher HOMA-IR value that is associated with
increased visceral adiposity, triglycerides (TG), total
cholesterol/high-density lipoprotein cholesterol (HDL-C), TG/HDL-C, plasma
insulin, GSK-3, corticosterone and decreased 17β-oestradiol. However, these
effects were attenuated in OVX+COC, OVX+SPL and OVX+COC+SPL rats compared to
OVX rats. OVX rats had lower PAI-1 than SHM rats, whereas the beneficial
effect on IR and other parameters by COC or SPL was accompanied with
increased PAI-1. Improvement of IR and other parameters with combined COC
and SPL in OVX rats was accompanied with reduced PAI-1. Conclusion: Taken together, COC or SPL improves IR independent of PAI-1, whereas a
combination of COC and SPL in OVX rats ameliorates IR in a PAI-1-dependent
manner.
Collapse
Affiliation(s)
- Adeyanju Oluwaseun Aremu
- HOPE Cardiometabolic Research Team and Department of Physiology, University of Ilorin, Nigeria.,Cardiometabolic Research Unit, Department of Physiology, College of Medicine and Health Sciences, Afe Babalola University, Nigeria
| | - Dibia Chinaza Lilian
- HOPE Cardiometabolic Research Team and Department of Physiology, University of Ilorin, Nigeria
| | - Soladoye Ayodele Olufemi
- Cardiometabolic Research Unit, Department of Physiology, College of Health sciences, Bowen University, Nigeria
| | | |
Collapse
|
22
|
Burke SJ, Batdorf HM, Huang TY, Jackson JW, Jones KA, Martin TM, Rohli KE, Karlstad MD, Sparer TE, Burk DH, Campagna SR, Noland RC, Soto PL, Collier JJ. One week of continuous corticosterone exposure impairs hepatic metabolic flexibility, promotes islet β-cell proliferation, and reduces physical activity in male C57BL/6 J mice. J Steroid Biochem Mol Biol 2019; 195:105468. [PMID: 31536768 PMCID: PMC6939671 DOI: 10.1016/j.jsbmb.2019.105468] [Citation(s) in RCA: 12] [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: 05/11/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 10/26/2022]
Abstract
Clinical glucocorticoid use, and diseases that produce elevated circulating glucocorticoids, promote drastic changes in body composition and reduction in whole body insulin sensitivity. Because steroid-induced diabetes is the most common form of drug-induced hyperglycemia, we investigated mechanisms underlying the recognized phenotypes associated with glucocorticoid excess. Male C57BL/6 J mice were exposed to either 100ug/mL corticosterone (cort) or vehicle in their drinking water. Body composition measurements revealed an increase in fat mass with drastically reduced lean mass during the first week (i.e., seven days) of cort exposure. Relative to the vehicle control group, mice receiving cort had a significant reduction in insulin sensitivity (measured by insulin tolerance test) five days after drug intervention. The increase in insulin resistance significantly correlated with an increase in the number of Ki-67 positive β-cells. Moreover, the ability to switch between fuel sources in liver tissue homogenate substrate oxidation assays revealed reduced metabolic flexibility. Furthermore, metabolomics analyses revealed a decrease in liver glycolytic metabolites, suggesting reduced glucose utilization, a finding consistent with onset of systemic insulin resistance. Physical activity was reduced, while respiratory quotient was increased, in mice receiving corticosterone. The majority of metabolic changes were reversed upon cessation of the drug regimen. Collectively, we conclude that changes in body composition and tissue level substrate metabolism are key components influencing the reductions in whole body insulin sensitivity observed during glucocorticoid administration.
Collapse
Affiliation(s)
- Susan J Burke
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States
| | - Heidi M Batdorf
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States
| | - Tai-Yu Huang
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States
| | - Joseph W Jackson
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, United States
| | - Katarina A Jones
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States
| | - Thomas M Martin
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Kristen E Rohli
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, United States
| | - Michael D Karlstad
- Department of Surgery, University of Tennessee Health Science Center, Knoxville, TN 37920, United States
| | - Tim E Sparer
- Department of Microbiology, University of Tennessee, Knoxville, TN 37996, United States
| | - David H Burk
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States
| | - Shawn R Campagna
- Department of Chemistry, University of Tennessee, Knoxville, TN 37996, United States
| | - Robert C Noland
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States
| | - Paul L Soto
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States; Department of Psychology, Louisiana State University, Baton Rouge, LA 70803, United States
| | - J Jason Collier
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, United States; Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, United States.
| |
Collapse
|
23
|
Adeyanju OA, Olatunji LA. Drospirenone-containing oral contraceptives do not affect glucose regulation and circulating corticosterone. J Basic Clin Physiol Pharmacol 2019; 30:jbcpp-2018-0184. [PMID: 31469652 DOI: 10.1515/jbcpp-2018-0184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 06/17/2019] [Indexed: 12/19/2022]
Abstract
Background Combined oral contraceptive (COC) use has been associated with an increased risk of insulin resistance (IR) and other adverse cardiovascular events, despite efforts to reduce the dosage and/or progestin type. COC containing drospirenone (DRSP) is an analog of spironolactone, hence its antimineralocorticoid and antiandrogenic characteristics have been deemed beneficial, although the benefits and/or negative outcome of its usage have not been fully elucidated. We therefore hypothesized that COC with DRSP component will not affect glucose regulation and circulating corticosterone. Method Ten-week-old female Wistar rats were divided into three groups: control (CON), ethinylestradiol/drospirenone COC (EE/DRSP)-treated, and ethinylestradiol/levonorgestrel COC (EE/LN)-treated rats. The treatment lasted for 8 weeks. Results Results showed that with the exception of lipid profiles, EE/LN but not EE/DRSP COC treatment affected body weight, glucose tolerance, plasma insulin, corticosterone, (IR), and pancreatic β-cell dysfunction. Conclusion Taken together, the findings showed that the beneficial effect of EE/DRSP could possibly be through the DRSP component. The result also implies that COCs containing DRSP may be a better and safer means of contraception than those with LN with less cardiovascular risks.
Collapse
Affiliation(s)
- Oluwaseun A Adeyanju
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin 240003, Nigeria.,Cardiometabolic Research Unit, Department of Physiology, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Lawrence A Olatunji
- Department of Physiology, College of Health Sciences, University of Ilorin, PMB 1515, Ilorin 240003, Nigeria, Phone: +2348035755360
| |
Collapse
|
24
|
Akbib S, Stichelmans J, Stangé G, Ling Z, Assefa Z, Hellemans KH. Glucocorticoids and checkpoint tyrosine kinase inhibitors stimulate rat pancreatic beta cell proliferation differentially. PLoS One 2019; 14:e0212210. [PMID: 30779812 PMCID: PMC6380609 DOI: 10.1371/journal.pone.0212210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cell therapy for diabetes could benefit from the identification of small-molecule compounds that increase the number of functional pancreatic beta cells. Using a newly developed screening assay, we previously identified glucocorticoids as potent stimulators of human and rat beta cell proliferation. We now compare the stimulatory action of these steroid hormones to a selection of checkpoint tyrosine kinase inhibitors that were also found to activate the cell cycle-in beta cells and analyzed their respective effects on DNA-synthesis, beta cell numbers and expression of cell cycle regulators. Our data using glucocorticoids in combination with a receptor antagonist, mifepristone, show that 48h exposure is sufficient to allow beta cells to pass the cell cycle restriction point and to become committed to cell division regardless of sustained glucocorticoid-signaling. To reach the end-point of mitosis another 40h is required. Within 14 days glucocorticoids stimulate up to 75% of the cells to undergo mitosis, which indicates that these steroid hormones act as proliferation competence-inducing factors. In contrast, by correlating thymidine-analogue incorporation to changes in absolute cell numbers, we show that the checkpoint kinase inhibitors, as compared to glucocorticoids, stimulate DNA-synthesis only during a short time-window in a minority of cells, insufficient to give a measurable increase of beta cell numbers. Glucocorticoids, but not the kinase inhibitors, were also found to induce changes in the expression of checkpoint regulators. Our data, using checkpoint kinase-specific inhibitors further point to a role for Chk1 and Cdk1 in G1/S transition and progression of beta cells through the cell cycle upon stimulation with glucocorticoids.
Collapse
Affiliation(s)
- Sarah Akbib
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Jordy Stichelmans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Geert Stangé
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Zhidong Ling
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Beta Cell Bank, University Hospital Brussels, Brussels, Belgium
| | - Zerihun Assefa
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
| | - Karine H. Hellemans
- Unit Diabetes Pathology and Therapy, Diabetes Research Cluster, Vrije Universiteit Brussel, Brussels, Belgium
- Center for Beta Cell Therapy in Diabetes, Brussels, Belgium
| |
Collapse
|
25
|
Mo Z, Li L, Yu H, Wu Y, Li H. Coumarins ameliorate diabetogenic action of dexamethasone via Akt activation and AMPK signaling in skeletal muscle. J Pharmacol Sci 2019; 139:151-157. [PMID: 30733181 DOI: 10.1016/j.jphs.2019.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 12/29/2018] [Accepted: 01/09/2019] [Indexed: 02/06/2023] Open
Abstract
Glucocorticoids are widely prescribed for lots of pathological conditions, however, can produce 'Cushingoid' side effects including central obesity, glucose intolerance, insulin resistance and so forth. Our study is intended to investigate the improving effects of coumarins on diabetogenic action of dexamethasone in vivo and in vitro and elucidate potential mechanisms. ICR mice treated with dexamethasone for 21 days exhibited decreased body weight, increased blood glucose and impaired glucose tolerance, which were prevented by fraxetin (40 mg/kg/day), esculin (40 mg/kg/day) and osthole (20 mg/kg/day), respectively. Esculin, fraxetin and osthole also could promote glucose uptake in normal C2C12 myotubes, and improve insulin resistance in myotubes induced by dexamethasone. Western blotting results indicated that esculin, fraxetin and osthole could boost Akt activation, stimulate GLUT4 translocation, thus alleviate insulin resistance. Esculin and osthole also could activate AMPK, thereby phosphorylate TBC1D1 at Ser237, and consequently ameliorate diabetogenic action of dexamethasone. Our study indicates coumarins as potential anti-diabetic candidates or leading compounds for drug development.
Collapse
Affiliation(s)
- Zejun Mo
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, People's Republic of China
| | - Linghuan Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Haiwen Yu
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Yingqi Wu
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China
| | - Hanbing Li
- Institute of Pharmacology, Zhejiang University of Technology, Hangzhou 310014, People's Republic of China; Section of Endocrinology, School of Medicine, Yale University, New Haven 06520, USA.
| |
Collapse
|
26
|
Hemmer MC, Wierer M, Schachtrup K, Downes M, Hübner N, Evans RM, Uhlenhaut NH. E47 modulates hepatic glucocorticoid action. Nat Commun 2019; 10:306. [PMID: 30659202 PMCID: PMC6338785 DOI: 10.1038/s41467-018-08196-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 12/17/2018] [Indexed: 01/24/2023] Open
Abstract
Glucocorticoids (GCs) are effective drugs, but their clinical use is compromised by severe side effects including hyperglycemia, hyperlipidemia and obesity. They bind to the Glucocorticoid Receptor (GR), which acts as a transcription factor. The activation of metabolic genes by GR is thought to underlie these adverse effects. We identify the bHLH factor E47 as a modulator of GR target genes. Using mouse genetics, we find that E47 is required for the regulation of hepatic glucose and lipid metabolism by GR, and that loss of E47 prevents the development of hyperglycemia and hepatic steatosis in response to GCs. Here we show that E47 and GR co-occupy metabolic promoters and enhancers. E47 is needed for the efficient recruitment of GR and coregulators such as Mediator to chromatin. Altogether, our results illustrate how GR and E47 regulate hepatic metabolism, and might provide an entry point for novel therapies with reduced side effects. Glucocorticoids (GCs) are widely used anti-inflammatory drugs; however, long-term treatment causes metabolic side effects. Here, the authors show that E47 is a modulator of glucocorticoid receptor activity for a subset of target genes in mouse liver, and that loss of E47 protects mice from hyperglycemia and hepatic steatosis in response to GCs.
Collapse
Affiliation(s)
- M Charlotte Hemmer
- Molecular Endocrinology, Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), IDO, Ingolstädter Landstr. 1, 85764, Neuherberg, Munich, Germany
| | - Michael Wierer
- Department of Proteomics and Signal Transduction, Max Planck Institute for Biochemistry, Am Klopferspitz 18, 82152, Martinsried, Germany
| | - Kristina Schachtrup
- Center for Chronic Immunodeficiency, University Medical Center and Faculty of Biology, University of Freiburg, 79106, Freiburg, Germany
| | - Michael Downes
- The Salk Institute for Biological Studies & HHMI, 10010 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences & DZHK (German Center for Cardiovascular Research), Charité-Universitätsmedizin & Berlin Institute of Health (BIH), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle Strasse 10, 13125, Berlin, Germany
| | - Ronald M Evans
- The Salk Institute for Biological Studies & HHMI, 10010 N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - N Henriette Uhlenhaut
- Molecular Endocrinology, Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), IDO, Ingolstädter Landstr. 1, 85764, Neuherberg, Munich, Germany. .,The Salk Institute for Biological Studies & HHMI, 10010 N Torrey Pines Rd, La Jolla, CA, 92037, USA. .,Cardiovascular and Metabolic Sciences & DZHK (German Center for Cardiovascular Research), Charité-Universitätsmedizin & Berlin Institute of Health (BIH), Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle Strasse 10, 13125, Berlin, Germany. .,Gene Center, Ludwig-Maximilians-Universität München (LMU), Feodor-Lynen-Straße 25, 81377, Munich, Germany.
| |
Collapse
|
27
|
Courty E, Besseiche A, Do TTH, Liboz A, Aguid FM, Quilichini E, Buscato M, Gourdy P, Gautier JF, Riveline JP, Haumaitre C, Buyse M, Fève B, Guillemain G, Blondeau B. Adaptive β-Cell Neogenesis in the Adult Mouse in Response to Glucocorticoid-Induced Insulin Resistance. Diabetes 2019; 68:95-108. [PMID: 30327384 DOI: 10.2337/db17-1314] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 10/11/2018] [Indexed: 11/13/2022]
Abstract
Both type 1 and type 2 diabetes are characterized by deficient insulin secretion and decreased β-cell mass. Thus, regenerative strategies to increase β-cell mass need to be developed. To characterize mechanisms of β-cell plasticity, we studied a model of severe insulin resistance in the adult mouse and defined how β-cells adapt. Chronic corticosterone (CORT) treatment was given to adult mice and led to rapid insulin resistance and adaptive increased insulin secretion. Adaptive and massive increase of β-cell mass was observed during treatment up to 8 weeks. β-Cell mass increase was partially reversible upon treatment cessation and reinduced upon subsequent treatment. β-Cell neogenesis was suggested by an increased number of islets, mainly close to ducts, and increased Sox9 and Ngn3 mRNA levels in islets, but lineage-tracing experiments revealed that neoformed β-cells did not derive from Sox9- or Ngn3-expressing cells. CORT treatment after β-cell depletion partially restored β-cells. Finally, β-cell neogenesis was shown to be indirectly stimulated by CORT because serum from CORT-treated mice increased β-cell differentiation in in vitro cultures of pancreatic buds. Altogether, the results present a novel model of β-cell neogenesis in the adult mouse and identify the presence of neogenic factors in the serum of CORT-treated mice.
Collapse
Affiliation(s)
- Emilie Courty
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Paris, France
- Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Adrien Besseiche
- Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, Paris, France
| | - Thi Thu Huong Do
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Paris, France
- Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Alexandrine Liboz
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Paris, France
- Hospitalo-Universitary Institute, ICAN, Paris, France
| | | | - Evans Quilichini
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Paris, France
| | - Melissa Buscato
- Institute of Metabolic and Cardiovascular Diseases, UMR1048, INSERM, UPS, Université de Toulouse, Toulouse, France
| | - Pierre Gourdy
- Institute of Metabolic and Cardiovascular Diseases, UMR1048, INSERM, UPS, Université de Toulouse, Toulouse, France
- Service de Diabétologie, CHU de Toulouse, Toulouse, France
| | - Jean-François Gautier
- Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, Paris, France
- Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Department of Diabetes and Endocrinology, University Paris-Diderot 7, Sorbonne Paris Cité, Paris, France
| | - Jean-Pierre Riveline
- Sorbonne Université, INSERM, Centre de Recherche des Cordeliers, Paris, France
- Lariboisière Hospital, Assistance Publique-Hôpitaux de Paris, Department of Diabetes and Endocrinology, University Paris-Diderot 7, Sorbonne Paris Cité, Paris, France
| | - Cécile Haumaitre
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, Paris, France
| | - Marion Buyse
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Paris, France
- Hospitalo-Universitary Institute, ICAN, Paris, France
- Université Paris-Sud, EA 4123, Chatenay-Malabry, France
- Department of Pharmacy, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Bruno Fève
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Paris, France
- Hospitalo-Universitary Institute, ICAN, Paris, France
- Department of Endocrinology, Saint-Antoine Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ghislaine Guillemain
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Paris, France
- Hospitalo-Universitary Institute, ICAN, Paris, France
| | - Bertrand Blondeau
- Sorbonne Université, INSERM, Saint-Antoine Research Center, Paris, France
- Hospitalo-Universitary Institute, ICAN, Paris, France
| |
Collapse
|
28
|
Zheng X, Bi W, Yang G, Zhao J, Wang J, Li X, Zhou X. Hyperglycemia Induced by Chronic Restraint Stress in Mice Is Associated With Nucleus Tractus Solitarius Injury and Not Just the Direct Effect of Glucocorticoids. Front Neurosci 2018; 12:983. [PMID: 30618599 PMCID: PMC6305899 DOI: 10.3389/fnins.2018.00983] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 12/07/2018] [Indexed: 12/20/2022] Open
Abstract
Chronic restraint stress (CRS) can affect hypothalamic-pituitary-adrenal (HPA) axis activity and increase glucocorticoid levels. Glucocorticoids are stress hormones that regulate multiple aspects of energy homeostasis. Stress also impairs glucose tolerance. The aim of this study was to investigate the cause of insulin-resistant hyperglycemia during CRS. We produced the CRS models (a 7-day restraint followed by a 3-day free moving procedure, total of 4 cycles for 40 days) in mice, detected the parameters related to glucose metabolism, and compared them to those of the dexamethasone (DEX) injection (0.2 mg/kg i.p., also a 4 cycle procedure as the CRS). The results showed that the CRS induced a moderate (not higher than 11 mmol/L) and irreversible insulin-resistant hyperglycemia in about 1/3 of the individuals, and all the restrained mice had adrenal hypertrophy. CRS induced the apoptosis of neurons in the anterior part of commissural subnucleus of nucleus tractus solitarius (acNTS) in the hyperglycemic mice, and acNTS mechanical damage also led to insulin-resistant hyperglycemia. In contrast, in the DEX-treated mice, adrenal gland atrophy was evident. The glucose and insulin tolerance varied with the delay of determination. DEX exposure in vivo does not induce the apoptosis of neurons in NTS. This study indicates that restraint stress and DEX induce metabolic disorders through different mechanisms. During CRS, injury (apoptosis) of glucose-sensitive acNTS neurons cause dysregulation of blood glucose. This study also suggests the mouse restraint stress model has value as a potential application in the study of stress-induced hyperglycemia.
Collapse
Affiliation(s)
- Xiang Zheng
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Wenjie Bi
- Department of Anatomy, Histology and Embryology, Chengdu Medical College, Chengdu, China
| | - Guizhi Yang
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jia Zhao
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| | - Jie Wang
- MOE Key Laboratory of Bioinformatics, Bioinformatics Division, Center for Synthetic and System Biology, Department of Automation, Tsinghua University, Beijing, China
| | - Xiaojing Li
- Department of Histology and Embryology, Fuzhou Medical College, Nanchang University, Fuzhou, China
| | - Xue Zhou
- Department of Histology, Embryology and Neurobiology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
29
|
Omolekulo TE, Areola ED, Badmus OO, Michael OS, Kim I, Olatunji LA. Inhibition of adenosine deaminase and xanthine oxidase by valproic acid abates hepatic triglyceride accumulation independent of corticosteroids in female rats treated with estrogen-progestin. Can J Physiol Pharmacol 2018; 96:1092-1103. [PMID: 30001502 DOI: 10.1139/cjpp-2018-0231] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elevated circulating uric acid has been postulated to play an important pathophysiological role in estrogen-progestin combined oral contraceptive (COC)-induced hypertension and endothelial dysfunction. We hypothesized that disruption of glucoregulation and liver triglyceride (TG) accumulation induced by COC use would be abated by valproic acid (VPA) treatment through suppression of adenosine deaminase (ADA) and xanthine oxidase (XO) activities. Female Wistar rats aged 9-10 weeks were treated with a combination of estrogen-progestin COC steroids (1.0 μg ethinylestradiol and 5.0 μg levonorgestrel; p.o.) with or without VPA (100.0 mg/kg; p.o.) daily for 6 weeks. The result shows that the disrupted glucoregulation and associated elevated hepatic ADA activity, plasma and hepatic XO activity, uric acid (UA), TG/HDL-cholesterol, total cholesterol, and malondialdehyde induced by COC treatment were attenuated by VPA treatment. However, VPA did not have any effect on plasma aldosterone, corticosterone, ADA, circulating and hepatic free fatty acid. Our results demonstrate that suppression of plasma and hepatic XO activities, along with hepatic ADA activity and UA by VPA treatment, protects against disrupted glucoregulation and increased liver TG by COC independent of elevated corticosteroids. The findings imply that VPA would provide protection against the development of cardiometabolic disorder via inhibition of the ADA/XO/UA-mediated pathway.
Collapse
Affiliation(s)
- Tolulope Eniola Omolekulo
- a HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Emmanuel Damilare Areola
- a HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olufunto Olayinka Badmus
- a HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,b Department of Public Health, Kwara State University, Malete, Nigeria
| | - Olugbenga Samuel Michael
- a HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria.,c Cardiometabolic Research Unit, Department of Physiology, College of Health Sciences, Bowen University, Iwo, Nigeria
| | - Inkyeom Kim
- d Cardiovascular Research Institute and Department of Pharmacology, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Lawrence Aderemi Olatunji
- a HOPE Cardiometabolic Research Team, Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
30
|
Adeyanju OA, Soetan OA, Soladoye AO, Olatunji LA. Oral hormonal therapy with ethinylestradiol–levonorgestrel improves insulin resistance, obesity, and glycogen synthase kinase-3 independent of circulating mineralocorticoid in estrogen-deficient rats. Can J Physiol Pharmacol 2018; 96:577-586. [DOI: 10.1139/cjpp-2017-0630] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Estrogen deficiency has been associated with increased incidence of cardiovascular diseases , and recent clinical trials of standard formulations of hormonal therapies have not demonstrated consistent beneficial effects. Estrogen–progestin therapy has been used as exogenous estrogen to normalize depressed estrogen level during menopause. Ovariectomized rodents mimic an estrogen-deficient state in that they develop cardiometabolic dysfunction, including insulin resistance (IR). We therefore hypothesized that hormonal therapy with combined oral contraceptive steroids, ethinylestradiol–levonorgestrel (EEL), improves IR, obesity, and glycogen synthase kinase-3 (GSK-3) through reduction of circulating mineralocorticoid in ovariectomized rats. Twelve-week-old female Wistar rats were divided into 4 groups: sham-operated (SHM) and ovariectomized (OVX) rats were treated with or without EEL (1.0 μg ethinylestradiol and 5.0 μg levonorgestrel) daily for 8 weeks. Results showed that OVX or SHM + EEL treated rats had increased HOMA-IR (homeostatic model assessment of IR), 1 h postload glucose, HOMA-β, triglycerides (TG), total cholesterol (TC), TC/HDL cholesterol, TG/HDL cholesterol, plasma insulin, GSK-3, corticosterone, and aldosterone. On the other hand, OVX + EEL treatment ameliorated all these effects except that of aldosterone. Taken together, the results demonstrate that oral hormonal replacement with EEL improves IR and pancreatic β-cell function and suppresses GSK-3 and glucocorticoid independent of circulating aldosterone, suggesting a positive cardiometabolic effect of oral EEL therapy in estrogen-deficient rats.
Collapse
Affiliation(s)
- Oluwaseun A. Adeyanju
- Cardiovascular Research Laboratory, Department of Physiology, University of Ilorin, Ilorin, Nigeria
- Cardiometabolic Research Unit, Department of Physiology, Afe Babalola University, Ado-Ekiti, Ekiti State, Nigeria
| | - Olaniyi A. Soetan
- Cardiovascular Research Laboratory, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| | - Ayodele O. Soladoye
- Cardiovascular Research Laboratory, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A. Olatunji
- Cardiovascular Research Laboratory, Department of Physiology, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
31
|
Su WJ, Peng W, Gong H, Liu YZ, Zhang Y, Lian YJ, Cao ZY, Wu R, Liu LL, Wang B, Wang YX, Jiang CL. Antidiabetic drug glyburide modulates depressive-like behavior comorbid with insulin resistance. J Neuroinflammation 2017; 14:210. [PMID: 29084550 PMCID: PMC5663104 DOI: 10.1186/s12974-017-0985-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Abundant reports indicated that depression was often comorbid with type 2 diabetes and even metabolic syndrome. Considering they might share common biological origins, it was tentatively attributed to the chronic cytokine-mediated inflammatory response which was induced by dysregulation of HPA axis and overactivation of innate immunity. However, the exact mechanisms remain obscure. Herein, we mainly focused on the function of the NLRP3 inflammasome to investigate this issue. METHODS Male C57BL/6 mice were subjected to 12 weeks of chronic unpredictable mild stress (CUMS), some of which were injected with glyburide or fluoxetine. After CUMS procedure, behavioral and metabolic tests were carried out. In order to evaluate the systemic inflammation associated with inflammasome activation, IL-1β and inflammasome components in hippocampi and pancreases, as well as corticosterone and IL-1β in serum were detected separately. Moreover, immunostaining was performed to assess morphologic characteristics of pancreases. RESULTS In the present study, we found that 12 weeks' chronic stress resulted in depressive-like behavior comorbid with insulin resistance. Furthermore, antidiabetic drug glyburide, an inhibitor of the NLRP3 inflammasome, was discovered to be effective in preventing the experimental comorbidity. In brief, it improved behavioral performance, ameliorated insulin intolerance as well as insulin signaling in the hippocampus possibly through inhibiting NLRP3 inflammasome activation by suppressing the expression of TXNIP. CONCLUSIONS All these evidence supported our hypothesis that chronic stress led to comorbidity of depressive-like behavior and insulin resistance via long-term mild inflammation. More importantly, based on the beneficial effects of blocking the activation of the NLRP3 inflammasome, we provided a potential therapeutic target for clinical comorbidity and a new strategy for management of both diabetes and depression.
Collapse
Affiliation(s)
- Wen-Jun Su
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Wei Peng
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Hong Gong
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yun-Zi Liu
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yi Zhang
- Department of Psychiatry, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yong-Jie Lian
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Zhi-Yong Cao
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
- Department of Psychiatry, The 102nd Hospital of PLA, 55 North Heping Road, Changzhou, China
| | - Ran Wu
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Lin-Lin Liu
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Bo Wang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Yun-Xia Wang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| | - Chun-Lei Jiang
- Laboratory of Stress Medicine, Faculty of Psychology and Mental Health, Second Military Medical University, 800 Xiangyin Road, Shanghai, China
| |
Collapse
|
32
|
Abstract
Psychological stress is common in many physical illnesses and is increasingly recognized as a risk factor for disease onset and progression. An emerging body of literature suggests that stress has a role in the aetiology of type 2 diabetes mellitus (T2DM) both as a predictor of new onset T2DM and as a prognostic factor in people with existing T2DM. Here, we review the evidence linking T2DM and psychological stress. We highlight the physiological responses to stress that are probably related to T2DM, drawing on evidence from animal work, large epidemiological studies and human laboratory trials. We discuss population and clinical studies linking psychological and social stress factors with T2DM, and give an overview of intervention studies that have attempted to modify psychological or social factors to improve outcomes in people with T2DM.
Collapse
Affiliation(s)
- Ruth A Hackett
- Department of Behavioural Science and Health, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
| | - Andrew Steptoe
- Department of Behavioural Science and Health, University College London, 1-19 Torrington Place, London WC1E 6BT, UK
| |
Collapse
|
33
|
Impact of Fish Oil Supplementation and Interruption of Fructose Ingestion on Glucose and Lipid Homeostasis of Rats Drinking Different Concentrations of Fructose. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4378328. [PMID: 28929113 PMCID: PMC5591931 DOI: 10.1155/2017/4378328] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 06/20/2017] [Accepted: 06/28/2017] [Indexed: 12/28/2022]
Abstract
Background. Continuous fructose consumption may cause elevation of circulating triacylglycerol. However, how much of this alteration is reverted after the removal of fructose intake is not known. We explored this question and compared the efficacy of this approach with fish oil supplementation. Methods. Male Wistar rats were divided into the following groups: control (C), fructose (F) (water intake with 10% or 30% fructose for 9 weeks), fish oil (FO), and fructose/fish oil (FFO). Fish oil was supplemented only for the last 33 days of fructose ingestion. Half of the F group remained for additional 8 weeks without fructose ingestion (FR). Results. Fructose ingestion reduced food intake to compensate for the increased energy obtained through water ingestion, independent of fructose concentration. Fish oil supplementation exerted no impact on these parameters, but the removal of fructose from water recovered both ingestion behaviors. Plasma triacylglycerol augmented significantly during the second and third weeks (both fructose groups). Fish oil supplementation did not attenuate the elevation in triacylglycerol caused by fructose intake, but the interruption of sugar consumption normalized this parameter. Conclusion. Elevation in triacylglyceridemia may be recovered by removing fructose from diet, suggesting that it is never too late to repair improper dietary habits.
Collapse
|
34
|
Oral Corticosterone Administration Reduces Insulitis but Promotes Insulin Resistance and Hyperglycemia in Male Nonobese Diabetic Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:614-626. [PMID: 28061324 DOI: 10.1016/j.ajpath.2016.11.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/03/2016] [Accepted: 11/23/2016] [Indexed: 12/11/2022]
Abstract
Steroid-induced diabetes is the most common form of drug-induced hyperglycemia. Therefore, metabolic and immunological alterations associated with chronic oral corticosterone were investigated using male nonobese diabetic mice. Three weeks after corticosterone delivery, there was reduced sensitivity to insulin action measured by insulin tolerance test. Body composition measurements revealed increased fat mass and decreased lean mass. Overt hyperglycemia (>250 mg/dL) manifested 6 weeks after the start of glucocorticoid administration, whereas 100% of the mice receiving the vehicle control remained normoglycemic. This phenotype was fully reversed during the washout phase and readily reproducible across institutions. Relative to the vehicle control group, mice receiving corticosterone had a significant enhancement in pancreatic insulin-positive area, but a marked decrease in CD3+ cell infiltration. In addition, there were striking increases in both citrate synthase gene expression and enzymatic activity in skeletal muscle of mice in the corticosterone group relative to vehicle control. Moreover, glycogen synthase expression was greatly enhanced, consistent with elevations in muscle glycogen storage in mice receiving corticosterone. Corticosterone-induced hyperglycemia, insulin resistance, and changes in muscle gene expression were all reversed by the end of the washout phase, indicating that the metabolic alterations were not permanent. Thus, male nonobese diabetic mice allow for translational studies on the metabolic and immunological consequences of glucocorticoid-associated interventions in a mouse model with genetic susceptibility to autoimmune disease.
Collapse
|
35
|
Gasparini SJ, Weber MC, Henneicke H, Kim S, Zhou H, Seibel MJ. Continuous corticosterone delivery via the drinking water or pellet implantation: A comparative study in mice. Steroids 2016; 116:76-82. [PMID: 27815034 DOI: 10.1016/j.steroids.2016.10.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/26/2016] [Indexed: 12/22/2022]
Abstract
In order to investigate the effects of glucocorticoid excess in rodent models, reliable methods of continuous glucocorticoid delivery are essential. The current study compares two methods of corticosterone (CS) delivery in regards to their ability to induce typical adverse outcomes such as fat accrual, insulin resistance, sarcopenia and bone loss. Eight-week-old mice received CS for 4weeks either via the drinking water (25-100μgCS/mL) or through weekly surgical implantation of slow release pellets containing 1.5mg CS. Both methods induced abnormal fat mass accrual, inhibited lean mass accretion and bone expansion, suppressed serum osteocalcin levels and induced severe insulin resistance. There was a clear dose dependant relationship between the CS concentrations in the drinking water and the severity of the phenotype, with a concentration of 50μg CS/mL drinking water most closely matching the metabolic changes induced by weekly pellet implantations. In contrast to pellets, however, delivery of CS via the drinking water resulted in a consistent diurnal exposure pattern, closely mimicking the kinetics of clinical glucocorticoid therapy. In addition, the method is safe, inexpensive, easily adjustable, non-invasive and avoids operative stress to the animals. Our data demonstrate that delivery of CS via the drinking water has advantages over weekly implantations of slow-release pellets. A dose of 50μg CS/mL drinking water is appropriate for the investigation of chronic glucocorticoid excess in mice.
Collapse
Affiliation(s)
- Sylvia J Gasparini
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia.
| | - Marie-Christin Weber
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia; Department of Rheumatology and Clinical Immunology, Charité University Hospital, Berlin, Germany
| | - Holger Henneicke
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia; DFG-Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
| | - Sarah Kim
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia
| | - Hong Zhou
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia
| | - Markus J Seibel
- Bone Research Program, ANZAC Research Institute, The University of Sydney, Sydney, Australia; Concord Medical School, The University of Sydney, Sydney, Australia.
| |
Collapse
|
36
|
Li YC, Liu YM, Shen JD, Chen JJ, Pei YY, Fang XY. Resveratrol Ameliorates the Depressive-Like Behaviors and Metabolic Abnormalities Induced by Chronic Corticosterone Injection. Molecules 2016; 21:molecules21101341. [PMID: 27754387 PMCID: PMC6274283 DOI: 10.3390/molecules21101341] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/24/2016] [Accepted: 10/06/2016] [Indexed: 12/17/2022] Open
Abstract
Chronic glucocorticoid exposure is known to cause depression and metabolic disorders. It is critical to improve abnormal metabolic status as well as depressive-like behaviors in patients with long-term glucocorticoid therapy. This study aimed to investigate the effects of resveratrol on the depressive-like behaviors and metabolic abnormalities induced by chronic corticosterone injection. Male ICR mice were administrated corticosterone (40 mg/kg) by subcutaneous injection for three weeks. Resveratrol (50 and 100 mg/kg), fluoxetine (20 mg/kg) and pioglitazone (10 mg/kg) were given by oral gavage 30 min prior to corticosterone administration. The behavioral tests showed that resveratrol significantly reversed the depressive-like behaviors induced by corticosterone, including the reduced sucrose preference and increased immobility time in the forced swimming test. Moreover, resveratrol also increased the secretion of insulin, reduced serum level of glucose and improved blood lipid profiles in corticosterone-treated mice without affecting normal mice. However, fluoxetine only reverse depressive-like behaviors, and pioglitazone only prevent the dyslipidemia induced by corticosterone. Furthermore, resveratrol and pioglitazone decreased serum level of glucagon and corticosterone. The present results indicated that resveratrol can ameliorate depressive-like behaviors and metabolic abnormalities induced by corticosterone, which suggested that the multiple effects of resveratrol could be beneficial for patients with depression and/or metabolic syndrome associated with long-term glucocorticoid therapy.
Collapse
Affiliation(s)
- Yu-Cheng Li
- College of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China.
| | - Ya-Min Liu
- College of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China.
| | - Ji-Duo Shen
- College of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China.
| | - Jun-Jie Chen
- College of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China.
| | - Yang-Yi Pei
- College of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China.
| | - Xiao-Yan Fang
- College of Pharmacy, Henan University of Traditional Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
37
|
Wong SK, Chin KY, Suhaimi FH, Fairus A, Ima-Nirwana S. Animal models of metabolic syndrome: a review. Nutr Metab (Lond) 2016; 13:65. [PMID: 27708685 PMCID: PMC5050917 DOI: 10.1186/s12986-016-0123-9] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 09/10/2016] [Indexed: 01/11/2023] Open
Abstract
Metabolic syndrome (MetS) consists of several medical conditions that collectively predict the risk for cardiovascular disease better than the sum of individual conditions. The risk of developing MetS in human depends on synergy of both genetic and environmental factors. Being a multifactorial condition with alarming rate of prevalence nowadays, establishment of appropriate experimental animal models mimicking the disease state in humans is crucial in order to solve the difficulties in evaluating the pathophysiology of MetS in human. This review aims to summarize the underlying mechanisms involved in the pathophysiology of dietary, genetic, and pharmacological models of MetS. Furthermore, we will discuss the usefulness, suitability, pros and cons of these animal models. Even though numerous animal models of MetS have been established, further investigations on the invention of new animal model and clarification of plausible mechanisms are still necessary to confer a better understanding to researchers on the selection of animal models for their studies.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaakob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaakob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur Malaysia
| | - Farihah Hj Suhaimi
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaakob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur Malaysia
| | - Ahmad Fairus
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaakob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaakob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur Malaysia
| |
Collapse
|
38
|
Abstract
Carbohydrate, lipid, and protein metabolism are largely controlled by the interplay of various hormones, which includes those secreted by the pancreatic islets of Langerhans. While typically representing only 1% to 2% of the total pancreatic mass, the islets have a remarkable ability to adapt to disparate situations demanding a change in hormone release, such as peripheral insulin resistance. There are many different routes to the onset of insulin resistance, including obesity, lipodystrophy, glucocorticoid excess, and the chronic usage of atypical antipsychotic drugs. All of these situations are coupled to an increase in pancreatic islet size, often with a corresponding increase in insulin production. These adaptive responses within the islets are ultimately intended to maintain glycemic control and to promote macronutrient homeostasis during times of stress. Herein, we review the consequences of specific metabolic trauma that lead to insulin resistance and the corresponding adaptive alterations within the pancreatic islets.
Collapse
Affiliation(s)
- Susan J. Burke
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808
| | - Michael D. Karlstad
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Knoxville, TN 37920
| | - J. Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA 70808
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Knoxville, TN 37920
| |
Collapse
|
39
|
Impact of Glucocorticoid Excess on Glucose Tolerance: Clinical and Preclinical Evidence. Metabolites 2016; 6:metabo6030024. [PMID: 27527232 PMCID: PMC5041123 DOI: 10.3390/metabo6030024] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 07/28/2016] [Accepted: 07/29/2016] [Indexed: 12/28/2022] Open
Abstract
Glucocorticoids (GCs) are steroid hormones that exert important physiological actions on metabolism. Given that GCs also exert potent immunosuppressive and anti-inflammatory actions, synthetic GCs such as prednisolone and dexamethasone were developed for the treatment of autoimmune- and inflammatory-related diseases. The synthetic GCs are undoubtedly efficient in terms of their therapeutic effects, but are accompanied by significant adverse effects on metabolism, specifically glucose metabolism. Glucose intolerance and reductions in insulin sensitivity are among the major concerns related to GC metabolic side effects, which may ultimately progress to type 2 diabetes mellitus. A number of pre-clinical and clinical studies have aimed to understand the repercussions of GCs on glucose metabolism and the possible mechanisms of GC action. This review intends to summarize the main alterations that occur in liver, skeletal muscle, adipose tissue, and pancreatic islets in the context of GC-induced glucose intolerance. For this, both experimental (animals) and clinical studies were selected and, whenever possible, the main cellular mechanisms involved in such GC-side effects were discussed.
Collapse
|
40
|
Diz-Chaves Y, Gil-Lozano M, Toba L, Fandiño J, Ogando H, González-Matías LC, Mallo F. Stressing diabetes? The hidden links between insulinotropic peptides and the HPA axis. J Endocrinol 2016; 230:R77-94. [PMID: 27325244 DOI: 10.1530/joe-16-0118] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 06/20/2016] [Indexed: 12/25/2022]
Abstract
Diabetes mellitus exerts metabolic stress on cells and it provokes a chronic increase in the long-term activity of the hypothalamus-pituitary-adrenocortical (HPA) axis, perhaps thereby contributing to insulin resistance. GLP-1 receptor (GLP-1R) agonists are pleiotropic hormones that not only affect glycaemic and metabolic control, but they also produce many other effects including activation of the HPA axis. In fact, several of the most relevant effects of GLP-1 might involve, at least in part, the modulation of the HPA axis. Thus, the anorectic activity of GLP-1 could be mediated by increasing CRF at the hypothalamic level, while its lipolytic effects could imply a local increase in glucocorticoids and glucocorticoid receptor (GC-R) expression in adipose tissue. Indeed, the potent activation of the HPA axis by GLP-1R agonists occurs within the range of therapeutic doses and with a short latency. Interestingly, the interactions of GLP-1 with the HPA axis may underlie most of the effects of GLP-1 on food intake control, glycaemic metabolism, adipose tissue biology and the responses to stress. Moreover, such activity has been observed in animal models (mice and rats), as well as in normal humans and in type I or type II diabetic patients. Accordingly, better understanding of how GLP-1R agonists modulate the activity of the HPA axis in diabetic subjects, especially obese individuals, will be crucial to design new and more efficient therapies for these patients.
Collapse
Affiliation(s)
- Yolanda Diz-Chaves
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Manuel Gil-Lozano
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Laura Toba
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Juan Fandiño
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Hugo Ogando
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Lucas C González-Matías
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| | - Federico Mallo
- Laboratory of EndocrinologyCenter for Biomedical Research - CINBIO, University of Vigo, Vigo, Spain Instituto de Investigación Sanitaria Galicia Sur - IISGSVigo, Spain
| |
Collapse
|
41
|
Regulation of Glucose Homeostasis by Glucocorticoids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015. [PMID: 26215992 DOI: 10.1007/978-1-4939-2895-8_5] [Citation(s) in RCA: 412] [Impact Index Per Article: 41.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids are steroid hormones that regulate multiple aspects of glucose homeostasis. Glucocorticoids promote gluconeogenesis in liver, whereas in skeletal muscle and white adipose tissue they decrease glucose uptake and utilization by antagonizing insulin response. Therefore, excess glucocorticoid exposure causes hyperglycemia and insulin resistance. Glucocorticoids also regulate glycogen metabolism. In liver, glucocorticoids increase glycogen storage, whereas in skeletal muscle they play a permissive role for catecholamine-induced glycogenolysis and/or inhibit insulin-stimulated glycogen synthesis. Moreover, glucocorticoids modulate the function of pancreatic α and β cells to regulate the secretion of glucagon and insulin, two hormones that play a pivotal role in the regulation of blood glucose levels. Overall, the major glucocorticoid effect on glucose homeostasis is to preserve plasma glucose for brain during stress, as transiently raising blood glucose is important to promote maximal brain function. In this chapter we will discuss the current understanding of the mechanisms underlying different aspects of glucocorticoid-regulated mammalian glucose homeostasis.
Collapse
|
42
|
Rafacho A, Ortsäter H, Nadal A, Quesada I. Glucocorticoid treatment and endocrine pancreas function: implications for glucose homeostasis, insulin resistance and diabetes. J Endocrinol 2014; 223:R49-62. [PMID: 25271217 DOI: 10.1530/joe-14-0373] [Citation(s) in RCA: 142] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Glucocorticoids (GCs) are broadly prescribed for numerous pathological conditions because of their anti-inflammatory, antiallergic and immunosuppressive effects, among other actions. Nevertheless, GCs can produce undesired diabetogenic side effects through interactions with the regulation of glucose homeostasis. Under conditions of excess and/or long-term treatment, GCs can induce peripheral insulin resistance (IR) by impairing insulin signalling, which results in reduced glucose disposal and augmented endogenous glucose production. In addition, GCs can promote abdominal obesity, elevate plasma fatty acids and triglycerides, and suppress osteocalcin synthesis in bone tissue. In response to GC-induced peripheral IR and in an attempt to maintain normoglycaemia, pancreatic β-cells undergo several morphofunctional adaptations that result in hyperinsulinaemia. Failure of β-cells to compensate for this situation favours glucose homeostasis disruption, which can result in hyperglycaemia, particularly in susceptible individuals. GC treatment does not only alter pancreatic β-cell function but also affect them by their actions that can lead to hyperglucagonaemia, further contributing to glucose homeostasis imbalance and hyperglycaemia. In addition, the release of other islet hormones, such as somatostatin, amylin and ghrelin, is also affected by GC administration. These undesired GC actions merit further consideration for the design of improved GC therapies without diabetogenic effects. In summary, in this review, we consider the implication of GC treatment on peripheral IR, islet function and glucose homeostasis.
Collapse
Affiliation(s)
- Alex Rafacho
- Department of Physiological SciencesCenter of Biological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, BrazilDepartment of Clinical Science and EducationSödersjukhuset, Karolinska Institutet, SE-11883 Stockholm, SwedenInstitute of Bioengineering and the Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM)Miguel Hernández University, University Avenue s/n, 03202, Elche, Spain
| | - Henrik Ortsäter
- Department of Physiological SciencesCenter of Biological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, BrazilDepartment of Clinical Science and EducationSödersjukhuset, Karolinska Institutet, SE-11883 Stockholm, SwedenInstitute of Bioengineering and the Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM)Miguel Hernández University, University Avenue s/n, 03202, Elche, Spain
| | - Angel Nadal
- Department of Physiological SciencesCenter of Biological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, BrazilDepartment of Clinical Science and EducationSödersjukhuset, Karolinska Institutet, SE-11883 Stockholm, SwedenInstitute of Bioengineering and the Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM)Miguel Hernández University, University Avenue s/n, 03202, Elche, Spain
| | - Ivan Quesada
- Department of Physiological SciencesCenter of Biological Sciences, Federal University of Santa Catarina (UFSC), 88040-900, Florianópolis, SC, BrazilDepartment of Clinical Science and EducationSödersjukhuset, Karolinska Institutet, SE-11883 Stockholm, SwedenInstitute of Bioengineering and the Biomedical Research Center in Diabetes and Associated Metabolic Disorders (CIBERDEM)Miguel Hernández University, University Avenue s/n, 03202, Elche, Spain
| |
Collapse
|
43
|
Fransson L, Rosengren V, Saha TK, Grankvist N, Islam T, Honkanen RE, Sjöholm Å, Ortsäter H. Mitogen-activated protein kinases and protein phosphatase 5 mediate glucocorticoid-induced cytotoxicity in pancreatic islets and β-cells. Mol Cell Endocrinol 2014; 383:126-36. [PMID: 24361515 DOI: 10.1016/j.mce.2013.12.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 12/15/2013] [Accepted: 12/16/2013] [Indexed: 11/21/2022]
Abstract
Glucocorticoid excess is associated with glucose intolerance and diabetes. In addition to inducing insulin resistance, glucocorticoids impair β-cell function and cause β-cell apoptosis. In this study we show that dexamethasone activates mitogen-activated protein kinases (MAPKs) signaling in MIN6 β-cells, as evident by enhanced phosphorylation of p38 MAPK and c-Jun N-terminal kinase (JNK). In contrast, the integrated stress response pathway was inhibited by dexamethasone. A p38 MAPK inhibitor attenuated dexamethasone-induced apoptosis in β-cells and isolated islets and decreased glucocorticoid receptor phosphorylation at S220. In contrast, a JNK inhibitor augmented DNA fragmentation and dexamethasone-induced formation of cleaved caspase 3. We also show that inhibition of protein phosphatase 5 (PP5) augments apoptosis in dexamethasone-exposed islets and β-cells, with a concomitant activation of p38 MAPK. In conclusion, our data provide evidence that in islets and β-cells, p38 MAPK and JNK phosphorylation work in concert with PP5 to regulate the cytotoxic effects exerted by glucocorticoids.
Collapse
Affiliation(s)
- Liselotte Fransson
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Victoria Rosengren
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Titu Kumar Saha
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Nina Grankvist
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Tohidul Islam
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Richard E Honkanen
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Department of Internal Medicine, Södertälje Hospital, SE 152 86 Södertälje, Sweden
| | - Åke Sjöholm
- Department of Internal Medicine, Södertälje Hospital, SE 152 86 Södertälje, Sweden; Department of Biochemistry and Molecular Biology, University of South Alabama, College of Medicine, Mobile, AL, USA
| | - Henrik Ortsäter
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Research Unit, Södertälje Hospital, SE-152 86 Södertälje, Sweden.
| |
Collapse
|
44
|
Fransson L, dos Santos C, Wolbert P, Sjöholm Å, Rafacho A, Ortsäter H. Liraglutide counteracts obesity and glucose intolerance in a mouse model of glucocorticoid-induced metabolic syndrome. Diabetol Metab Syndr 2014; 6:3. [PMID: 24423471 PMCID: PMC3905931 DOI: 10.1186/1758-5996-6-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 01/09/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Glucocorticoid excess is commonly associated with diabetogenic effects, including insulin resistance and glucose intolerance. The effects of the long-term glucagon-like peptide 1 receptor agonist treatment on the metabolic syndrome-like conditions are not yet fully elucidated. Thus, we aimed to test whether long-term liraglutide treatment could be effective as a therapy to counteract the metabolic dysfunctions induced by chronic glucocorticoid exposure. METHODS Mice were given corticosterone or vehicle via their drinking water for five consecutive weeks. In addition, mice were treated with once-daily injections of either PBS or liraglutide. RESULTS Liraglutide treatment slowed progression towards obesity and ectopic fat deposition in liver that otherwise occurred in corticosterone-treated mice. The drug reduced the increment in serum insulin caused by corticosterone, but did not affect the reduction of insulin sensitivity. Furthermore, liraglutide improved glucose control in mice exposed to corticosterone as evident by a delay in the progression towards post-prandial hyperglycemia and enhanced glucose clearance during a glucose tolerance test. Glucose-stimulated C-peptide levels were higher in those mice that had received liraglutide and corticosterone compared to mice that had been treated with corticosterone alone, indicating a positive role of liraglutide for beta-cell function. Morphometric analysis revealed increased beta- and alpha-cell masses that were associated with more Ki67-positive islet cells in corticosterone-treated mice irrespective of whether they were co-treated with liraglutide or not. Liraglutide had no discernible effect on alpha-cell mass. CONCLUSION Liraglutide can be beneficial for subjects at risk of developing metabolic complications as a result of glucocorticoid excess.
Collapse
Affiliation(s)
- Liselotte Fransson
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Research Center Floor 3, 118 83, Stockholm, Sweden
| | - Cristiane dos Santos
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Petra Wolbert
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Research Center Floor 3, 118 83, Stockholm, Sweden
| | - Åke Sjöholm
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Research Center Floor 3, 118 83, Stockholm, Sweden
- Department of Internal Medicine, Södertälje Hospital, SE-152 86, Södertälje, Sweden
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Alex Rafacho
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Henrik Ortsäter
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Research Center Floor 3, 118 83, Stockholm, Sweden
- Research Unit, Södertälje Hospital, SE-152 86, Södertälje, Sweden
| |
Collapse
|