1
|
Ye J, Ye H, Wang M, Zhao Y. Total serum IL-6 and TNF-C levels in children with bronchopneumonia following treatment with methylprednisolone in combination with azithromycin. Am J Transl Res 2021; 13:9458-9464. [PMID: 34540066 PMCID: PMC8430067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/18/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To analyze the expression levels of total serum interleukin (IL)-6 and tumor necrosis factor (TNF)-C in children with bronchopneumonia treated by methylprednisolone in combination with azithromycin. METHODS Eighty-three children with bronchopneumonia were randomly divided into a test group (TG) and a control group (CG). The TG was comprised of 40 children treated with methylprednisolone combined with azithromycin, whereas the CG was comprised of 43 patients who received methylprednisolone monotherapy. The post-treatment effective rates and occurrence of adverse reactions were compared between the two groups. In addition, the resolution times of symptoms such as fever, cough, moist rale, asthma, and shadow on the lung X-ray were recorded. The levels of the inflammatory factors tumor necrosis factor-C (TNF-C) and interleukin-6 (IL-6) were measured after treatment. The quality of life was evaluated and compared based on the Medical Outcome Study (MOS) 36-Item Short-Form Health Survey (SF-36). RESULTS The total effective rate in the TG was significantly higher than that in the CG. The expression levels of TNF-C and IL-6 in the TG were significantly lower than those in the CG. The resolution times of the clinical symptoms were significantly shorter in the TG than in the CG. The ACT (Asthma Control Test) score in the TG was significantly lower than that in the CG. The TG presented with a significantly lower incidence of adverse reactions than that the CG. CONCLUSION The combined administration of methylprednisolone and antibiotics can effectively improve the levels of serum inflammatory factors and the clinical symptoms in children with bronchopneumonia.
Collapse
Affiliation(s)
- Jianya Ye
- School of Nursing, Hebei University of Chinese MedicineShijiazhuang, Hebei, China
| | - Haiyan Ye
- Department of Obstetrics, Shenzhou HospitalShenzhou, Hengshui, China
| | - Minghui Wang
- School of Nursing, Hebei University of Chinese MedicineShijiazhuang, Hebei, China
| | - Yanjun Zhao
- Department of Pharmacy, The First Affiliated Hospital of Zhejiang Chinese Medical UniversityHangzhou, Zhejiang, China
| |
Collapse
|
2
|
Mavroudis PD, Jusko WJ. Mathematical modeling of mammalian circadian clocks affecting drug and disease responses. J Pharmacokinet Pharmacodyn 2021; 48:375-386. [PMID: 33725238 DOI: 10.1007/s10928-021-09746-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/26/2021] [Indexed: 12/28/2022]
Abstract
To align with daily environmental changes, most physiological processes in mammals exhibit a time-of-day rhythmicity. This circadian control of physiology is intrinsically driven by a cell-autonomous clock gene network present in almost all cells of the body that drives rhythmic expression of genes that regulate numerous molecular and cellular processes. Accordingly, many aspects of pharmacology and toxicology also oscillate in a time-of-day manner giving rise to diverse effects on pharmacokinetics and pharmacodynamics. Genome-wide studies and mathematical modeling are available tools that have significantly improved our understanding of these nonlinear aspects of physiology and therapeutics. In this manuscript current literature and our prior work on the model-based approaches that have been used to explore circadian genomic systems of mammals are reviewed. Such basic understanding and having an integrative approach may provide new strategies for chronotherapeutic drug treatments and yield new insights for the restoration of the circadian system when altered by diseases.
Collapse
Affiliation(s)
- Panteleimon D Mavroudis
- Quantitative Pharmacology, DMPK, Sanofi, Waltham, MA, 02451, USA. .,State University of New York, School of Pharmacy and Pharmaceutical Sciences, University of Buffalo, Buffalo, NY, USA.
| | - William J Jusko
- State University of New York, School of Pharmacy and Pharmaceutical Sciences, University of Buffalo, Buffalo, NY, USA
| |
Collapse
|
3
|
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
|
4
|
Ayyar VS, Jusko WJ. Transitioning from Basic toward Systems Pharmacodynamic Models: Lessons from Corticosteroids. Pharmacol Rev 2020; 72:414-438. [PMID: 32123034 DOI: 10.1124/pr.119.018101] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Technology in bioanalysis, -omics, and computation have evolved over the past half century to allow for comprehensive assessments of the molecular to whole body pharmacology of diverse corticosteroids. Such studies have advanced pharmacokinetic and pharmacodynamic (PK/PD) concepts and models that often generalize across various classes of drugs. These models encompass the "pillars" of pharmacology, namely PK and target drug exposure, the mass-law interactions of drugs with receptors/targets, and the consequent turnover and homeostatic control of genes, biomarkers, physiologic responses, and disease symptoms. Pharmacokinetic methodology utilizes noncompartmental, compartmental, reversible, physiologic [full physiologically based pharmacokinetic (PBPK) and minimal PBPK], and target-mediated drug disposition models using a growing array of pharmacometric considerations and software. Basic PK/PD models have emerged (simple direct, biophase, slow receptor binding, indirect response, irreversible, turnover with inactivation, and transduction models) that place emphasis on parsimony, are mechanistic in nature, and serve as highly useful "top-down" methods of quantitating the actions of diverse drugs. These are often components of more complex quantitative systems pharmacology (QSP) models that explain the array of responses to various drugs, including corticosteroids. Progressively deeper mechanistic appreciation of PBPK, drug-target interactions, and systems physiology from the molecular (genomic, proteomic, metabolomic) to cellular to whole body levels provides the foundation for enhanced PK/PD to comprehensive QSP models. Our research based on cell, animal, clinical, and theoretical studies with corticosteroids have provided ideas and quantitative methods that have broadly advanced the fields of PK/PD and QSP modeling and illustrates the transition toward a global, systems understanding of actions of diverse drugs. SIGNIFICANCE STATEMENT: Over the past half century, pharmacokinetics (PK) and pharmacokinetics/pharmacodynamics (PK/PD) have evolved to provide an array of mechanism-based models that help quantitate the disposition and actions of most drugs. We describe how many basic PK and PK/PD model components were identified and often applied to the diverse properties of corticosteroids (CS). The CS have complications in disposition and a wide array of simple receptor-to complex gene-mediated actions in multiple organs. Continued assessments of such complexities have offered opportunities to develop models ranging from simple PK to enhanced PK/PD to quantitative systems pharmacology (QSP) that help explain therapeutic and adverse CS effects. Concurrent development of state-of-the-art PK, PK/PD, and QSP models are described alongside experimental studies that revealed diverse CS actions.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| | - William J Jusko
- Department of Pharmaceutical Sciences University at Buffalo, School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| |
Collapse
|
5
|
Ayyar VS, DuBois DC, Nakamura T, Almon RR, Jusko WJ. Modeling Corticosteroid Pharmacokinetics and Pharmacodynamics, Part II: Sex Differences in Methylprednisolone Pharmacokinetics and Corticosterone Suppression. J Pharmacol Exp Ther 2019; 370:327-336. [PMID: 31197019 DOI: 10.1124/jpet.119.257527] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/10/2019] [Indexed: 12/23/2022] Open
Abstract
Methylprednisolone (MPL), a corticosteroid of intermediate potency, remains an important immunomodulatory agent for autoimmune diseases. Although sex differences in corticosteroid pharmacokinetics/pharmacodynamics (PK/PD) have been documented in humans, comprehensive preclinical assessments of such differences have not been conducted. Limited in vitro evidence indicates possible sex differences in corticosteroid PK and PD. Therefore, it is hypothesized that comparative PK/PD assessments of MPL disposition and selected PD actions in both sexes will provide insights into factors controlling sex differences in steroid responses. This report focused on the plasma and tissue pharmacokinetics of MPL and its adrenal suppressive effects. Because time-dependent (estrous) regulation of sex hormones in females can influence drug responses, female rats were studied in the proestrus (high estradiol/progesterone) and estrus (low estradiol/progesterone) phases of the reproductive cycle. Cohorts of male and female rats were given a 50 mg/kg bolus dose of MPL intramuscularly. Plasma and liver concentrations of MPL as well as plasma corticosterone concentrations were assayed using high-performance liquid chromatography. An enhanced minimal physiologically-based PK/PD model was developed to characterize MPL kinetics and corticosterone dynamics. The clearance of MPL was ∼3-fold higher in males compared with females, regardless of estrous phase, likely attributable to sex-specific hepatic metabolism in males. Strong inhibitory effects on adrenal suppression were observed in all animals. These temporal steroid profiles in plasma and tissues will be used to drive receptor/gene-mediated PD effects of MPL in both sexes, as described in a companion article (Part III). SIGNIFICANCE STATEMENT: Sex is a relevant factor influencing the pharmacokinetics (PK) and pharmacodynamics (PD) of drugs. Few preclinical PK/PD studies, however, include sex as a variable. Sex differences in the PK and adrenal suppressive effects of the synthetic corticosteroid, methylprednisolone, were assessed in male and female rats as a function of the 4-day rodent reproductive cycle. Drug exposure was 3-fold higher in females, regardless of estrous stage, compared with males. An extended minimal physiologically-based PK/PD model utilizing in vitro and in vivo measurements was developed and applied. These studies provide a framework to account for sex-dependent variability in drug and endogenous agonist (corticosterone) exposures, serving as a prelude to more intricate assessments of sex-related variability in receptor/gene-mediated PD corticosteroid actions.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., T.N., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York; and DMPK Research Department, Teijin Institute for Biomedical Research, Teijin Pharma, Tokyo, Japan (T.N.)
| | - Debra C DuBois
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., T.N., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York; and DMPK Research Department, Teijin Institute for Biomedical Research, Teijin Pharma, Tokyo, Japan (T.N.)
| | - Toshimichi Nakamura
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., T.N., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York; and DMPK Research Department, Teijin Institute for Biomedical Research, Teijin Pharma, Tokyo, Japan (T.N.)
| | - Richard R Almon
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., T.N., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York; and DMPK Research Department, Teijin Institute for Biomedical Research, Teijin Pharma, Tokyo, Japan (T.N.)
| | - William J Jusko
- Departments of Pharmaceutical Sciences (V.S.A., D.C.D., T.N., R.R.A., W.J.J.) and Biological Sciences (D.C.D., R.R.A.), State University of New York at Buffalo, Buffalo, New York; and DMPK Research Department, Teijin Institute for Biomedical Research, Teijin Pharma, Tokyo, Japan (T.N.)
| |
Collapse
|
6
|
Ekstrand C, Falkenö U, Kallings P, Tvedten H, Lilliehöök I. Plasma Dexamethasone Concentration in Relation to Glucose Response in the Horse. J Equine Vet Sci 2019. [DOI: 10.1016/j.jevs.2018.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
7
|
Ayyar VS, Sukumaran S, DuBois DC, Almon RR, Qu J, Jusko WJ. Receptor/gene/protein-mediated signaling connects methylprednisolone exposure to metabolic and immune-related pharmacodynamic actions in liver. J Pharmacokinet Pharmacodyn 2018; 45:557-575. [PMID: 29704219 DOI: 10.1007/s10928-018-9585-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 03/23/2018] [Indexed: 12/19/2022]
Abstract
A multiscale pharmacodynamic model was developed to characterize the receptor-mediated, transcriptomic, and proteomic determinants of corticosteroid (CS) effects on clinically relevant hepatic processes following a single dose of methylprednisolone (MPL) given to adrenalectomized (ADX) rats. The enhancement of tyrosine aminotransferase (TAT) mRNA, protein, and enzyme activity were simultaneously described. Mechanisms related to the effects of MPL on glucose homeostasis, including the regulation of CCAAT-enhancer binding protein-beta (C/EBPβ) and phosphoenolpyruvate carboxykinase (PEPCK) as well as insulin dynamics were evaluated. The MPL-induced suppression of circulating lymphocytes was modeled by coupling its effect on cell trafficking with pharmacogenomic effects on cell apoptosis via the hepatic (STAT3-regulated) acute phase response. Transcriptomic and proteomic time-course profiles measured in steroid-treated rat liver were utilized to model the dynamics of mechanistically relevant gene products, which were linked to associated systemic end-points. While time-courses of TAT mRNA, protein, and activity were well described by transcription-mediated changes, additional post-transcriptional processes were included to explain the lack of correlation between PEPCK mRNA and protein. The immune response model quantitatively discerned the relative roles of cell trafficking versus gene-mediated lymphocyte apoptosis by MPL. This systems pharmacodynamic model provides insights into the contributions of selected molecular events occurring in liver and explores mechanistic hypotheses for the multi-factorial control of clinically relevant pharmacodynamic outcomes.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Siddharth Sukumaran
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - Debra C DuBois
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA.,Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Richard R Almon
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA.,Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
8
|
Ayyar VS, Almon RR, DuBois DC, Sukumaran S, Qu J, Jusko WJ. Functional proteomic analysis of corticosteroid pharmacodynamics in rat liver: Relationship to hepatic stress, signaling, energy regulation, and drug metabolism. J Proteomics 2017; 160:84-105. [PMID: 28315483 DOI: 10.1016/j.jprot.2017.03.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 02/15/2017] [Accepted: 03/10/2017] [Indexed: 02/07/2023]
Abstract
Corticosteroids (CS) are anti-inflammatory agents that cause extensive pharmacogenomic and proteomic changes in multiple tissues. An understanding of the proteome-wide effects of CS in liver and its relationships to altered hepatic and systemic physiology remains incomplete. Here, we report the application of a functional pharmacoproteomic approach to gain integrated insight into the complex nature of CS responses in liver in vivo. An in-depth functional analysis was performed using rich pharmacodynamic (temporal-based) proteomic data measured over 66h in rat liver following a single dose of methylprednisolone (MPL). Data mining identified 451 differentially regulated proteins. These proteins were analyzed on the basis of temporal regulation, cellular localization, and literature-mined functional information. Of the 451 proteins, 378 were clustered into six functional groups based on major clinically-relevant effects of CS in liver. MPL-responsive proteins were highly localized in the mitochondria (20%) and cytosol (24%). Interestingly, several proteins were related to hepatic stress and signaling processes, which appear to be involved in secondary signaling cascades and in protecting the liver from CS-induced oxidative damage. Consistent with known adverse metabolic effects of CS, several rate-controlling enzymes involved in amino acid metabolism, gluconeogenesis, and fatty-acid metabolism were altered by MPL. In addition, proteins involved in the metabolism of endogenous compounds, xenobiotics, and therapeutic drugs including cytochrome P450 and Phase-II enzymes were differentially regulated. Proteins related to the inflammatory acute-phase response were up-regulated in response to MPL. Functionally-similar proteins showed large diversity in their temporal profiles, indicating complex mechanisms of regulation by CS. SIGNIFICANCE Clinical use of corticosteroid (CS) therapy is frequent and chronic. However, current knowledge on the proteome-level effects of CS in liver and other tissues is sparse. While transcriptomic regulation following methylprednisolone (MPL) dosing has been temporally examined in rat liver, proteomic assessments are needed to better characterize the tissue-specific functional aspects of MPL actions. This study describes a functional pharmacoproteomic analysis of dynamic changes in MPL-regulated proteins in liver and provides biological insight into how steroid-induced perturbations on a molecular level may relate to both adverse and therapeutic responses presented clinically.
Collapse
Affiliation(s)
- Vivaswath S Ayyar
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, NY, United States
| | - Richard R Almon
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, NY, United States; Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - Debra C DuBois
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, NY, United States; Department of Biological Sciences, State University of New York at Buffalo, Buffalo, NY, United States
| | - Siddharth Sukumaran
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, NY, United States
| | - Jun Qu
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, NY, United States
| | - William J Jusko
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, NY, United States.
| |
Collapse
|
9
|
Dogruk Unal A, Ayturk S, Aldemir D, Bascil Tutuncu N. Serum Adiponectin Level as a Predictor of Subclinical Cushing's Syndrome in Patients with Adrenal Incidentaloma. Int J Endocrinol 2016; 2016:8519362. [PMID: 27656211 PMCID: PMC5021502 DOI: 10.1155/2016/8519362] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/08/2016] [Accepted: 08/14/2016] [Indexed: 11/17/2022] Open
Abstract
Subclinical Cushing's syndrome (SCS) is a condition of slight but chronic cortisol excess in patients with adrenal incidentaloma (AI) without typical signs and symptoms of Cushing's syndrome. Adiponectin has potent roles in modulating energy balance and metabolic homeostasis and acts in opposition to glucocorticoids. This study aimed to evaluate adiponectin level in SCS and nonfunctional AI (NAI) patients and its relation with metabolic parameters. Patients with AI (n = 40) and metabolically healthy controls (n = 30) were included. In AI patients and controls, detailed medical history assessment, physical examinations, anthropometric measurements, and laboratory measurements were performed. Age, body mass index, waist circumference, and lipid profiles were significantly higher and waist-to-hip ratio and adiponectin level were significantly lower in the AI patients than in the controls. The midnight cortisol and urinary free cortisol levels were significantly higher in the SCS patients (n = 8) than in the NAI patients (n = 32). Adiponectin level of the SCS group was significantly lower than those of the NAI and control groups. The sensitivity and specificity for an adiponectin level of ≤13.00 ng/mL in predicting the presence of SCS were 87.5% and 77.4%, respectively. In conclusion, adiponectin is valuable in predicting the presence of SCS in AI patients.
Collapse
Affiliation(s)
- Asli Dogruk Unal
- Memorial Atasehir Hospital, Department of Endocrinology and Metabolism, Istanbul, Turkey
- *Asli Dogruk Unal:
| | - Semra Ayturk
- Trakya University Hospital, Department of Endocrinology and Metabolism, Edirne, Turkey
| | - Derya Aldemir
- Baskent University Hospital, Department of Biochemistry, Ankara, Turkey
| | | |
Collapse
|
10
|
Fang J, Sukumaran S, DuBois DC, Almon RR, Jusko WJ. Meta-modeling of methylprednisolone effects on glucose regulation in rats. PLoS One 2013; 8:e81679. [PMID: 24312573 PMCID: PMC3847111 DOI: 10.1371/journal.pone.0081679] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/15/2013] [Indexed: 01/01/2023] Open
Abstract
A retrospective meta-modeling analysis was performed to integrate previously reported data of glucocorticoid (GC) effects on glucose regulation following a single intramuscular dose (50 mg/kg), single intravenous doses (10, 50 mg/kg), and intravenous infusions (0.1, 0.2, 0.3 and 0.4 mg/kg/h) of methylprednisolone (MPL) in normal and adrenalectomized (ADX) male Wistar rats. A mechanistic pharmacodynamic (PD) model was developed based on the receptor/gene/protein-mediated GC effects on glucose regulation. Three major target organs (liver, white adipose tissue and skeletal muscle) together with some selected intermediate controlling factors were designated as important regulators involved in the pathogenesis of GC-induced glucose dysregulation. Assessed were dynamic changes of food intake and systemic factors (plasma glucose, insulin, free fatty acids (FFA) and leptin) and tissue-specific biomarkers (cAMP, phosphoenolpyruvate carboxykinase (PEPCK) mRNA and enzyme activity, leptin mRNA, interleukin 6 receptor type 1 (IL6R1) mRNA and Insulin receptor substrate-1 (IRS-1) mRNA) after acute and chronic dosing with MPL along with the GC receptor (GR) dynamics in each target organ. Upon binding to GR in liver, MPL dosing caused increased glucose production by stimulating hepatic cAMP and PEPCK activity. In adipose tissue, the rise in leptin mRNA and plasma leptin caused reduction of food intake, the exogenous source of glucose input. Down-regulation of IRS-1 mRNA expression in skeletal muscle inhibited the stimulatory effect of insulin on glucose utilization further contributing to hyperglycemia. The nuclear drug-receptor complex served as the driving force for stimulation or inhibition of downstream target gene expression within different tissues. Incorporating information such as receptor dynamics, as well as the gene and protein induction, allowed us to describe the receptor-mediated effects of MPL on glucose regulation in each important tissue. This advanced mechanistic model provides unique insights into the contributions of major tissues and quantitative hypotheses for the multi-factor control of a complex metabolic system.
Collapse
Affiliation(s)
- Jing Fang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Siddharth Sukumaran
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| | - Debra C. DuBois
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York, United States of America
| | - Richard R. Almon
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York, United States of America
| | - William J. Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, United States of America
| |
Collapse
|
11
|
Gao W, Jusko WJ. Modeling disease progression and rosiglitazone intervention in type 2 diabetic Goto-Kakizaki rats. J Pharmacol Exp Ther 2012; 341:617-25. [PMID: 22378938 DOI: 10.1124/jpet.112.192419] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The pharmacokinetics (PK) and pharmacodynamics (PD) of rosiglitazone were studied in type 2 diabetic (T2D) Goto-Kakizaki (GK) rats that received daily doses of 0, 5, or 10 mg/kg for 23 days followed by 60 days of washout. Blood glucose, plasma insulin, and hemoglobin A1c were determined over time. Oral glucose tolerance tests were performed before and at the end of treatment and after 20 days of washout to determine insulin sensitivity and β-cell function. Rosiglitazone effectively lowered glucose by inhibiting hepatic glucose production and enhancing insulin sensitivity. The glucose-insulin inter-regulation was characterized by a feedback model: glucose and insulin have their own production (k(in)) and elimination (k(out)) rate constants, whereas glucose stimulates insulin production (k(inI)) and insulin, in turn, promotes glucose utilization (k(outG)). Animal handling and placebo treatment affected glucose turnover with k(pl) = 0.388 kg/mg/day. The PK of rosiglitazone was fitted with a one-compartment model with first-order absorption. The effect of rosiglitazone was described as inhibition of k(inG) with I(max) = 0.296 and IC(50) = 1.97 μg/ml. Rosiglitazone also stimulated glucose utilization by improving insulin sensitivity with a linear factor S(R) = 0.0796 kg/mg. In GK rats, 23 days of treatment increased body weight but did not cause hemodilution. Weight gain was characterized with body weight input (k(s)(w)) and output (k(d)(w)), and rosiglitazone inhibited k(d)(w) with ID(50) = 96.8 mg/kg. The mechanistic PK/PD model quantitatively described the glucose-insulin system and body weights under chronic rosiglitazone treatment in T2D rats.
Collapse
Affiliation(s)
- Wei Gao
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | | |
Collapse
|
12
|
Gao W, Jusko WJ. Target-mediated pharmacokinetic and pharmacodynamic model of exendin-4 in rats, monkeys, and humans. Drug Metab Dispos 2012; 40:990-7. [PMID: 22338110 DOI: 10.1124/dmd.111.042291] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A mechanism-based pharmacokinetic-pharmacodynamic (PK/PD) model was developed for exendin-4 to account for receptor-mediated endocytosis via glucagon-like peptide 1 receptor (GLP-1R) as the primary mechanism for its nonlinear disposition. Time profiles of exendin-4 concentrations after intravenous, subcutaneous, and continuous intravenous infusion doses in rats, intravenous and subcutaneous doses in monkeys, and intravenous infusion and subcutaneous doses in humans were examined. Mean data for glucose and insulin after glucose challenges during exendin-4 treatment in healthy rats were analyzed. The PK model components included receptor binding, subsequent internalization and degradation, nonspecific tissue distribution, and linear first-order elimination from plasma. The absorption rate constant (k(a)) decreased with increasing doses in all three species. The clearance from the central compartment (CL(c)) (rats, 3.62 ml/min; monkeys, 2.39 ml · min(-1) · kg(-1); humans, 1.48 ml · min(-1) · kg(-1)) was similar to reported renal clearances. Selected PK parameters (CL(c), V(c), and k(off)) correlated allometrically with body weight. The equilibrium dissociation constant (K(D)) was within the reported range in rats (0.74 nM), whereas the value in monkeys (0.12 pM) was much lower than that in humans (1.38 nM). The effects of exendin-4 on the glucose-insulin system were described by a feedback model with a biphasic effect equation driven by free exendin-4 concentrations. Our generalized nonlinear PK/PD model for exendin-4 taking into account of drug binding to GLP-1R well described PK profiles after various routes of administration over a large range of doses in three species along with PD responses in healthy rats. The present model closely reflects underlying mechanisms of disposition and dynamics of exendin-4.
Collapse
Affiliation(s)
- Wei Gao
- 565 Hochstetter Hall, Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14260, USA
| | | |
Collapse
|
13
|
Abstract
Maintenance of energy metabolism and glucose homeostasis is achieved by the regulatory effects of many hormones and their interactions. Glucocorticoids produced from adrenal cortex and adiponectin produced by adipose tissue play important roles in the production, distribution, storage, and utilization of energy substrates. Glucocorticoids are involved in the activation of a number of catabolic processes by affecting the expression of a plethora of genes, while adiponectin acts primarily as an insulin sensitizer. Both are regulated by a number of physiological and pharmacological factors. Although the effects of glucocorticoids on adiponectin expression have been extensively studied in different in vitro, animal and clinical study settings, no consensus has been reached. This report reviews the primary literature concerning the effects of glucocorticoids on adiponectin expression and identifies potential reasons for the contradictory results between different studies. In addition, methods to gain better insights pertaining to the regulation of adiponectin expression are discussed.
Collapse
|
14
|
Fang J, DuBois DC, He Y, Almon RR, Jusko WJ. Dynamic modeling of methylprednisolone effects on body weight and glucose regulation in rats. J Pharmacokinet Pharmacodyn 2011; 38:293-316. [PMID: 21394487 DOI: 10.1007/s10928-011-9194-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Accepted: 02/14/2011] [Indexed: 12/21/2022]
Abstract
Influences of methylprednisolone (MPL) and food consumption on body weight (BW), and the effects of MPL on glycemic control including food consumption and the dynamic interactions among glucose, insulin, and free fatty acids (FFA) were evaluated in normal male Wistar rats. Six groups of animals received either saline or MPL via subcutaneous infusions at the rate of 0.03, 0.1, 0.2, 0.3 and 0.4 mg/kg/h for different treatment periods. BW and food consumption were measured twice a week. Plasma concentrations of MPL and corticosterone (CST) were determined at animal sacrifice. Plasma glucose, insulin, and FFA were measured at various times after infusion. Plasma MPL concentrations were simulated by a two-compartment model and used as the driving force in the pharmacodynamic (PD) analysis. All data were modeled using ADAPT 5. The MPL treatments caused reduction of food consumption and body weights in all dosing groups. The steroid also caused changes in plasma glucose, insulin, and FFA concentrations. Hyperinsulinemia was achieved rapidly at the first sampling time of 6 h; significant elevations of FFA were observed in all drug treatment groups; whereas only modest increases in plasma glucose were observed in the low dosing groups (0.03 and 0.1 mg/kg/h). Body weight changes were modeled by dual actions of MPL: inhibition of food consumption and stimulation of weight loss, with food consumption accounting for the input of energy for body weight. Dynamic models of glucose and insulin feedback interactions were extended to capture the major metabolic effects of FFA: stimulation of insulin secretion and inhibition of insulin-stimulated glucose utilization. These models of body weight and glucose regulation adequately captured the experimental data and reflect significant physiological interactions among glucose, insulin, and FFA. These mechanism-based PD models provide further insights into the multi-factor control of this essential metabolic system.
Collapse
Affiliation(s)
- Jing Fang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY 14260, USA
| | | | | | | | | |
Collapse
|
15
|
Gao W, Bihorel S, DuBois DC, Almon RR, Jusko WJ. Mechanism-based disease progression modeling of type 2 diabetes in Goto-Kakizaki rats. J Pharmacokinet Pharmacodyn 2010; 38:143-62. [PMID: 21127951 DOI: 10.1007/s10928-010-9182-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 11/04/2010] [Indexed: 11/25/2022]
Abstract
The dynamics of aging and type 2 diabetes (T2D) disease progression were investigated in normal [Wistar-Kyoto (WKY)] and diabetic [Goto-Kakizaki (GK)] rats and a mechanistic disease progression model was developed for glucose, insulin, and glycosylated hemoglobin (HbA1c) changes over time. The study included 30 WKY and 30 GK rats. Plasma glucose and insulin, blood glucose and HbA1c concentrations and hematological measurements were taken at ages 4, 8, 12, 16 and 20 weeks. A mathematical model described the development of insulin resistance (IR) and β-cell function with age/growth and diabetes progression. The model utilized transit compartments and an indirect response model to quantitate biomarker changes over time. Glucose, insulin and HbA1c concentrations in WKY rats increased to a steady-state at 8 weeks due to developmental changes. Glucose concentrations at 4 weeks in GK rats were almost twice those of controls, and increased to a steady-state after 8 weeks. Insulin concentrations at 4 weeks in GK rats were similar to controls, and then hyperinsulinemia occurred until 12-16 weeks of age indicating IR. Subsequently, insulin concentrations in GK rats declined to slightly below WKY controls due to β-cell failure. HbA1c showed a delayed increase relative to glucose. Modeling of HbA1c was complicated by age-related changes in hematology in rats. The diabetes model quantitatively described the glucose/insulin inter-regulation and HbA1c production and reflected the underlying pathogenic factors of T2D--IR and β-cell dysfunction. The model could be extended to incorporate other biomarkers and effects of various anti-diabetic drugs.
Collapse
Affiliation(s)
- Wei Gao
- Department of Pharmaceutical Sciences, State University of New York, 565 Hochstetter Hall, Buffalo, NY, USA
| | | | | | | | | |
Collapse
|