1
|
Abbas NAT, Fayed FA, El Sebaey RS, Hassan HA. Telmisartan and candesartan promote browning of white adipose tissue and reverse fatty liver changes in high fat diet fed male albino rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2359-2378. [PMID: 37831115 DOI: 10.1007/s00210-023-02771-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
Obesity is a key risk factor for many diseases, as cardiovascular disorders, diabetes, infertility, osteoarthritis, sleep apnea, non-alcoholic fatty liver disease (NAFLD) as well as increased risk for many cancers. Telmisartan and Candesartan cilexetil are angiotensin II receptor blockers which had proven to involve in pathogenesis of obesity and NAFLD. AIMS This work is designed to explore the possible mitigated effects of Telmisartan and Candesartan cilexetil on weight gain and fatty liver in high fat diet (HFD) fed rats. MAIN METHODS The HFD rat model was achieved with induction of NAFLD. For Seven weeks either telmisartan or candesartan were orally administered at doses of 5 and 10 mg/kg respectively once daily. The effects of both drugs were evaluated by measurements of rat's body weight, food intakes, length, body mass index (BMI), liver weight, inguinal and interscapular fat weights. In addition, we assayed lipid profile, liver functions tests, serum inflammatory cytokines, adipokine and leptin. Lastly, liver and adipose tissue histopathological structures were evaluated. KEY FINDINGS at end of experiment, telmisartan and candesartan were highly effective in decreasing rat's body weight from (213.1±2.68 to 191.2±2.54 and 203.5±5.89 gm , respectively), BMI, liver weight, fat weights in addition reduced serum levels of lipid and liver enzymes. Also, inflammatory cytokines were reduced with repaired histopathological insults in liver by significantly damped NAFLD score from (6.5 ±0.17 to 1±0 and 4 ±0, respectively) and decreased areas of adipocytes from (21239.12 to 5355.7 and 11607.1 um2 , respectively). SIGNIFICANCE Telmisartan and candesartan have therapeutic potential against obesity and NAFLD induced by HFD in rats. All the previous indices showed more improvement in telmisartan than candesartan group.
Collapse
Affiliation(s)
- Noha A T Abbas
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Fawkia A Fayed
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Rabab Saber El Sebaey
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Heba A Hassan
- Clinical Pharmacology Department, Faculty of Medicine, Zagazig University, Zagazig, 44519, Egypt.
- Department of Pharmacology, Faculty of Medicine, Mutah University, P.O. Box 7, Al-Karak, 61710, Jordan.
| |
Collapse
|
2
|
Malliou F, Andriopoulou CE, Gonzalez FJ, Kofinas A, Skaltsounis AL, Konstandi M. Oleuropein-Induced Acceleration of Cytochrome P450-Catalyzed Drug Metabolism: Central Role for Nuclear Receptor Peroxisome Proliferator-Activated Receptor α. Drug Metab Dispos 2021; 49:833-843. [PMID: 34162688 PMCID: PMC11022892 DOI: 10.1124/dmd.120.000302] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 04/06/2021] [Indexed: 11/22/2022] Open
Abstract
Oleuropein (OLE), the main constituent of Olea europaea, displays pleiotropic beneficial effects in health and disease, which are mainly attributed to its anti-inflammatory and cardioprotective properties. Several food supplements and herbal medicines contain OLE and are available without a prescription. This study investigated the effects of OLE on the main cytochrome P450s (P450s) catalyzing the metabolism of many prescribed drugs. Emphasis was given to the role of peroxisome proliferator-activated receptor α (PPARα), a nuclear transcription factor regulating numerous genes including P450s. 129/Sv wild-type and Ppara-null mice were treated with OLE for 6 weeks. OLE induced Cyp1a1, Cyp1a2, Cyp1b1, Cyp3a14, Cyp3a25, Cyp2c29, Cyp2c44, Cyp2d22, and Cyp2e1 mRNAs in liver of wild-type mice, whereas no similar effects were observed in Ppara-null mice, indicating that the OLE-induced effect on these P450s is mediated by PPARα. Activation of the pathways related to phosphoinositide 3-kinase/protein kinase B (AKT)/forkhead box protein O1, c-Jun N-terminal kinase, AKT/p70, and extracellular signal-regulated kinase participates in P450 induction by OLE. These data indicate that consumption of herbal medicines and food supplements containing OLE could accelerate the metabolism of drug substrates of the above-mentioned P450s, thus reducing their efficacy and the outcome of pharmacotherapy. Therefore, OLE-induced activation of PPARα could modify the effects of drugs due to their increased metabolism and clearance, which should be taken into account when consuming OLE-containing products with certain drugs, in particular those of narrow therapeutic window. SIGNIFICANCE STATEMENT: This study indicated that oleuropein, which belongs to the main constituents of the leaves and olive drupes of Olea europaea, induces the synthesis of the major cytochrome P450s (P450s) metabolizing the majority of prescribed drugs via activation of peroxisome proliferator-activated receptor α. This effect could modify the pharmacokinetic profile of co-administered drug substrates of the P450s, thus altering their therapeutic efficacy and toxicity.
Collapse
Affiliation(s)
- Foteini Malliou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (F.M., C.E.A., A.K., M.K.); Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Faculty of Pharmacy, School of Health Sciences, University of Athens, Athens, Greece (A.-L.S.)
| | - Christina E Andriopoulou
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (F.M., C.E.A., A.K., M.K.); Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Faculty of Pharmacy, School of Health Sciences, University of Athens, Athens, Greece (A.-L.S.)
| | - Frank J Gonzalez
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (F.M., C.E.A., A.K., M.K.); Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Faculty of Pharmacy, School of Health Sciences, University of Athens, Athens, Greece (A.-L.S.)
| | - Aristeidis Kofinas
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (F.M., C.E.A., A.K., M.K.); Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Faculty of Pharmacy, School of Health Sciences, University of Athens, Athens, Greece (A.-L.S.)
| | - Alexios-Leandros Skaltsounis
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (F.M., C.E.A., A.K., M.K.); Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Faculty of Pharmacy, School of Health Sciences, University of Athens, Athens, Greece (A.-L.S.)
| | - Maria Konstandi
- Department of Pharmacology, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece (F.M., C.E.A., A.K., M.K.); Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland (F.J.G.); and Faculty of Pharmacy, School of Health Sciences, University of Athens, Athens, Greece (A.-L.S.)
| |
Collapse
|
3
|
Lenahan C, Huang L, Travis ZD, Zhang JH. Scavenger Receptor Class B type 1 (SR-B1) and the modifiable risk factors of stroke. Chin Neurosurg J 2019; 5:30. [PMID: 32922929 PMCID: PMC7398188 DOI: 10.1186/s41016-019-0178-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/30/2019] [Indexed: 01/11/2023] Open
Abstract
Stroke is a devastating disease that occurs when a blood vessel in the brain is either blocked or ruptured, consequently leading to deficits in neurological function. Stroke consistently ranked as one of the top causes of mortality, and with the mean age of incidence decreasing, there is renewed interest to seek novel therapeutic treatments. The Scavenger Receptor Class B type 1 (SR-B1) is a multifunctional protein found on the surface of a variety of cells. Research has found that that SR-B1 primarily functions in an anti-inflammatory and anti-atherosclerotic capacity. In this review, we discuss the characteristics of SR-B1 and focus on its potential correlation with the modifiable risk factors of stroke. SR-B1 likely has an impact on stroke through its interaction with smoking, diabetes mellitus, diet, physical inactivity, obesity, hypercholesterolemia, atherosclerosis, coronary heart disease, hypertension, and sickle cell disease, all of which are critical risk factors in the pathogenesis of stroke.
Collapse
Affiliation(s)
- Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM 88003 USA
- Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA 92324 USA
| | - Lei Huang
- Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA 92324 USA
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA 92350 USA
- Department of Physiology & Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350 USA
| | - Zachary D. Travis
- Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA 92324 USA
- Department of Earth and Biological Sciences, School of Medicine, Loma Linda University, Loma Linda, CA 92350 USA
| | - John H. Zhang
- Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA 92324 USA
- Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, CA 92350 USA
- Department of Physiology & Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92350 USA
- Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, CA 92324 USA
| |
Collapse
|
4
|
Kaminski HJ, Himuro K, Alshaikh J, Gong B, Cheng G, Kusner LL. Differential RNA Expression Profile of Skeletal Muscle Induced by Experimental Autoimmune Myasthenia Gravis in Rats. Front Physiol 2016; 7:524. [PMID: 27891095 PMCID: PMC5102901 DOI: 10.3389/fphys.2016.00524] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/24/2016] [Indexed: 01/14/2023] Open
Abstract
The differential susceptibility of skeletal muscle by myasthenia gravis (MG) is not well understood. We utilized RNA expression profiling of extraocular muscle (EOM), diaphragm (DIA), and extensor digitorum (EDL) of rats with experimental autoimmune MG (EAMG) to evaluate the hypothesis that muscles respond differentially to injury produced by EAMG. EAMG was induced in female Lewis rats by immunization with acetylcholine receptor purified from the electric organ of the Torpedo. Six weeks later after rats had developed weakness and serum antibodies directed against the AChR, animals underwent euthanasia and RNA profiling performed on DIA, EDL, and EOM. Profiling results were validated by qPCR. Across the three muscles between the experiment and control groups, 359 probes (1.16%) with greater than 2-fold changes in expression in 7 of 9 series pairwise comparisons from 31,090 probes were identified with approximately two-thirds being increased. The three muscles shared 16 genes with increased expression and 6 reduced expression. Functional annotation demonstrated that these common expression changes fell predominantly into categories of metabolism, stress response, and signaling. Evaluation of specific gene function indicated that EAMG led to a change to oxidative metabolism. Genes related to muscle regeneration and suppression of immune response were activated. Evidence of a differential immune response among muscles was not evident. Each muscle had a distinct RNA profile but with commonality in gene categories expressed that are focused on muscle repair, moderation of inflammation, and oxidative metabolism.
Collapse
Affiliation(s)
- Henry J Kaminski
- Department of Neurology, George Washington University Washington, DC, USA
| | - Keiichi Himuro
- Department of Neurology, Graduate School of Medicine, Chiba University Chiba, Japan
| | - Jumana Alshaikh
- Department of Neurology, George Washington University Washington, DC, USA
| | - Bendi Gong
- Department of Pediatrics, Washington University St. Louis, MO, USA
| | - Georgiana Cheng
- Department of Pathobiology, Cleveland Clinic Cleveland, OH, USA
| | - Linda L Kusner
- Pharmacology and Physiology, George Washington University Washington, DC, USA
| |
Collapse
|
5
|
Chronic Moderate Alcohol Intakes Accelerate SR-B1 Mediated Reverse Cholesterol Transport. Sci Rep 2016; 6:33032. [PMID: 27618957 PMCID: PMC5020497 DOI: 10.1038/srep33032] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/18/2016] [Indexed: 11/17/2022] Open
Abstract
Cholesterol is essential for all animal life. However, a high level of cholesterol in the body is strongly associated with the progression of various severe diseases. In our study, the potential involvement of alcohol in the regulation of high density lipoprotein (HDL) receptor scavenger receptor class B and type I (SR-B1)-mediated reverse cholesterol transport was investigated. We separated male C57BL/6 mice into four diets: control, alcohol, Control + HC and alcohol + HC. The SR-B1 level and 1,1′-dioctadecyl-3,3,3′,3′-tetramethylindocarbocyanine perchlorate- high- density lipoprotein (DiI-HDL) uptake were also measured in AML12 cells and HL7702 cells treated with alcohol. The control + HC diet led to increased hepatic triglyceride and cholesterol levels while alcohol + HC led no significant change. Compared with that of the control group, the SR-B1 mRNA level was elevated by 27.1% (P < 0.05), 123.8% (P < 0.001) and 343.6% (P < 0.001) in the alcohol, control + HC and alcohol + HC groups, respectively. In AML12 and HL7702 cells, SR-B1 level and DiI-HDL uptake were repressed by SR-B1 siRNA or GW9662. However, these effects were reversed through alcohol treatment. These data suggest that a moderate amount of alcohol plays a novel role in reverse cholesterol transport, mainly mediated by PPARγ and SR-B1.
Collapse
|
6
|
Pereira VH, Marques F, Lages V, Pereira FG, Patchev A, Almeida OFX, Almeida-Palha J, Sousa N, Cerqueira JJ. Glucose intolerance after chronic stress is related with downregulated PPAR-γ in adipose tissue. Cardiovasc Diabetol 2016; 15:114. [PMID: 27538526 PMCID: PMC4990862 DOI: 10.1186/s12933-016-0433-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 08/06/2016] [Indexed: 01/09/2023] Open
Abstract
Background Chronic stress is associated with increased risk of glucose intolerance and cardiovascular diseases, albeit through undefined mechanisms. With the aim of gaining insights into the latter, this study examined the metabolic profile of young adult male rats that were exposed to chronic unpredictable stress. Methods Young adult male rats were submitted to 4 weeks of chronic unpredictable stress and allowed to recover for 5 weeks. An extensive analysis including of morphologic, biochemical and molecular parameters was carried out both after chronic unpredictable stress and after recovery from stress. Results After 28 days of chronic unpredictable stress (CUS) the animals submitted to this protocol displayed less weight gain than control animals. After 5 weeks of recovery the weight gain rebounded to similar values of controls. In addition, following CUS, fasting insulin levels were increased and were accompanied by signs of impaired glucose tolerance and elevated serum corticosteroid levels. This biochemical profile persisted into the post-stress recovery period, despite the restoration of baseline corticosteroid levels. The mRNA expression levels of peroxisome proliferator-activated receptor (PPAR)-γ and lipocalin-2 in white adipose tissue were, respectively, down- and up-regulated. Conclusions Reduction of PPAR-γ expression and generation of a pro-inflammatory environment by increased lipocalin-2 expression in white adipose tissue may contribute to stress-induced glucose intolerance.
Collapse
Affiliation(s)
- Vitor H Pereira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Vânia Lages
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipa G Pereira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | | | - Joana Almeida-Palha
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - João J Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
7
|
Sookoian S, Pirola CJ. Review: Genetics of the cardiometabolic syndrome: new insights and therapeutic implications. Ther Adv Cardiovasc Dis 2016; 1:37-47. [DOI: 10.1177/1753944707082702] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Although the definition of the phenotype is imprecise, cardiometabolic syndrome (CMS) includes a constellation of complex diseases such as type 2 diabetes, dislipidemias, central obesity and hypertension, proinflammatory and prothrombotic states, ovarian polycystosis and fatty liver. The genetics of each disease is complex in itself and varies in spectrum from monogenic and syndromic models of inheritance, usually rare, to the most common polygenic and multifactorial forms. In addition, human studies using the candidate-gene approach indicate that common genetic variants of several genes are associated with the development of CMS. Genome-wide scans have also provided several chromosomal regions associated with some of the components of CMS. In addition, through comparative genomics animal models can generate a map for candidate loci in humans and a promising approach is offered by bioinformatic tools for gene prioritization. Lastly, the involvement of genes whose products are already the targets for approved drugs, such as SLC6A4, PPARα and PPARγ , in the development of CMS suggests new avenues for CMS pharmacological treatment.
Collapse
Affiliation(s)
- Silvia Sookoian
- Departamento de Sustancias Vasoactivas y Cardiología Molecular, Instituto de Investigaciones A Lanari, Universidad de Buenos Aires-CONICET, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carlos J. Pirola
- Departamento de Sustancias Vasoactivas y Cardiología Molecular, Instituto de Investigaciones A Lanari, Universidad de Buenos Aires-CONICET, Ciudad Autónoma de Buenos Aires, Argentina, , pirola.carlos@lanari. fmed.uba.ar
| |
Collapse
|
8
|
Synthesis of Phthalimide Derivatives as Potential PPAR-γ Ligands. Mar Drugs 2016; 14:md14060112. [PMID: 27338418 PMCID: PMC4926071 DOI: 10.3390/md14060112] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 11/17/2022] Open
Abstract
Paecilocin A, a phthalide derivative isolated from the jellyfish-derived fungus Paecilomyces variotii, activates PPAR-γ (Peroxisome proliferator-activated receptor gamma) in rat liver Ac2F cells. Based on a SAR (Structure-activity relationships) study and in silico analysis of paecilocin A-mimetic derivatives, additional N-substituted phthalimide derivatives were synthesized and evaluated for PPAR-γ agonistic activity in both murine liver Ac2F cells and in human liver HepG2 cells by luciferase assay, and for adipogenic activity in 3T3-L1 cells. Docking simulation indicated PD6 was likely to bind most strongly to the ligand binding domain of PPAR-γ by establishing crucial H-bonds with key amino acid residues. However, in in vitro assays, PD1 and PD2 consistently displayed significant PPAR-γ activation in Ac2F and HepG2 cells, and adipogenic activity in 3T3-L1 preadipocytes.
Collapse
|
9
|
Watkins AM, Wood CR, Lin MT, Abbott BD. The effects of perfluorinated chemicals on adipocyte differentiation in vitro. Mol Cell Endocrinol 2015; 400:90-101. [PMID: 25448844 DOI: 10.1016/j.mce.2014.10.020] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 10/24/2014] [Accepted: 10/24/2014] [Indexed: 12/22/2022]
Abstract
The 3T3-L1 preadipocyte culture system has been used to examine numerous compounds that influence adipocyte differentiation or function. The perfluoroalkyl acids (PFAAs), used as surfactants in a variety of industrial applications, are of concern as environmental contaminants that are detected worldwide in human serum and animal tissues. This study was designed to evaluate the potential for PFAAs to affect adipocyte differentiation and lipid accumulation using mouse 3T3-L1 cells. Cells were treated with perfluorooctanoic acid (PFOA) (5-100 µM), perfluorononanoic acid (PFNA) (5-100 µM), perfluorooctane sulfonate (PFOS) (50-300 µM), perfluorohexane sulfonate (PFHxS) (40-250 µM), the peroxisome proliferator activated receptor (PPAR) PPARα agonist Wyeth-14,643 (WY-14,643), and the PPARγ agonist rosiglitazone. The PPARγ agonist was included as a positive control as this pathway is critical to adipocyte differentiation. The PPARα agonist was included as the PFAA compounds are known activators of this pathway. Cells were assessed morphometrically and biochemically for number, size, and lipid content. RNA was extracted for qPCR analysis of 13 genes selected for their importance in adipocyte differentiation and lipid metabolism. There was a significant concentration-related increase in cell number and decreased cell size after exposure to PFOA, PFHxS, PFOS, and PFNA. All four PFAA treatments produced a concentration-related decrease in the calculated average area occupied by lipid per cell. However, total triglyceride levels per well increased with a concentration-related trend for all compounds, likely due to the increased cell number. Expression of mRNA for the selected genes was affected by all exposures and the specific impacts depended on the particular compound and concentration. Acox1 and Gapdh were upregulated by all six compounds. The strongest overall effect was a nearly 10-fold induction of Scd1 by PFHxS. The sulfonated PFAAs produced numerous, strong changes in gene expression similar to the effects after treatment with the PPARγ agonist rosiglitazone. By comparison, the effects on gene expression were muted for the carboxylated PFAAs and for the PPARα agonist WY-14,643. In summary, all perfluorinated compounds increased cell number, decreased cell size, increased total triglyceride, and altered expression of genes associated with adipocyte differentiation and lipid metabolism.
Collapse
Affiliation(s)
- Andrew M Watkins
- Developmental Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Lab, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC 27709, USA.
| | - Carmen R Wood
- Developmental Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Lab, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC 27709, USA
| | - Mimi T Lin
- Developmental Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Lab, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC 27709, USA
| | - Barbara D Abbott
- Developmental Toxicology Branch, Toxicity Assessment Division, National Health and Environmental Effects Research Lab, Office of Research and Development, US Environmental Protection Agency, Research Triangle Park, NC 27709, USA
| |
Collapse
|
10
|
Konstandi M, Shah YM, Matsubara T, Gonzalez FJ. Role of PPARα and HNF4α in stress-mediated alterations in lipid homeostasis. PLoS One 2013; 8:e70675. [PMID: 23967086 PMCID: PMC3743822 DOI: 10.1371/journal.pone.0070675] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Accepted: 06/20/2013] [Indexed: 12/13/2022] Open
Abstract
Stress is a risk factor for several cardiovascular pathologies. PPARα holds a fundamental role in control of lipid homeostasis by directly regulating genes involved in fatty acid transport and oxidation. Importantly, PPARα agonists are effective in raising HDL-cholesterol and lowering triglycerides, properties that reduce the risk for cardiovascular diseases. This study investigated the role of stress and adrenergic receptor (AR)-related pathways in PPARα and HNF4α regulation and signaling in mice following repeated restraint stress or treatment with AR-antagonists administered prior to stress to block AR-linked pathways. Repeated restraint stress up-regulated Pparα and its target genes in the liver, including Acox, Acot1, Acot4, Cyp4a10, Cyp4a14 and Lipin2, an effect that was highly correlated with Hnf4α. In vitro studies using primary hepatocyte cultures treated with epinephrine or AR-agonists confirmed that hepatic AR/cAMP/PKA/CREB- and JNK-linked pathways are involved in PPARα and HNF4α regulation. Notably, restraint stress, independent of PPARα, suppressed plasma triglyceride levels. This stress-induced effect could be attributed in part to hormone sensitive lipase activation in the white adipose tissue, which was not prevented by the increased levels of perilipin. Overall, this study identifies a mechanistic basis for the modification of lipid homeostasis following stress and potentially indicates novel roles for PPARα and HNF4α in stress-induced lipid metabolism.
Collapse
Affiliation(s)
- Maria Konstandi
- Laboratory of Metabolism, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America.
| | | | | | | |
Collapse
|
11
|
Polyacetylenes from Notopterygium incisum--new selective partial agonists of peroxisome proliferator-activated receptor-gamma. PLoS One 2013; 8:e61755. [PMID: 23630612 PMCID: PMC3632601 DOI: 10.1371/journal.pone.0061755] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 03/12/2013] [Indexed: 12/22/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a key regulator of glucose and lipid metabolism and therefore an important pharmacological target to combat metabolic diseases. Since the currently used full PPARγ agonists display serious side effects, identification of novel ligands, particularly partial agonists, is highly relevant. Searching for new active compounds, we investigated extracts of the underground parts of Notopterygium incisum, a medicinal plant used in traditional Chinese medicine, and observed significant PPARγ activation using a PPARγ-driven luciferase reporter model. Activity-guided fractionation of the dichloromethane extract led to the isolation of six polyacetylenes, which displayed properties of selective partial PPARγ agonists in the luciferase reporter model. Since PPARγ activation by this class of compounds has so far not been reported, we have chosen the prototypical polyacetylene falcarindiol for further investigation. The effect of falcarindiol (10 µM) in the luciferase reporter model was blocked upon co-treatment with the PPARγ antagonist T0070907 (1 µM). Falcarindiol bound to the purified human PPARγ receptor with a Ki of 3.07 µM. In silico docking studies suggested a binding mode within the ligand binding site, where hydrogen bonds to Cys285 and Glu295 are predicted to be formed in addition to extensive hydrophobic interactions. Furthermore, falcarindiol further induced 3T3-L1 preadipocyte differentiation and enhanced the insulin-induced glucose uptake in differentiated 3T3-L1 adipocytes confirming effectiveness in cell models with endogenous PPARγ expression. In conclusion, we identified falcarindiol-type polyacetylenes as a novel class of natural partial PPARγ agonists, having potential to be further explored as pharmaceutical leads or dietary supplements.
Collapse
|
12
|
The Functions of PPARs in Aging and Longevity. PPAR Res 2011; 2007:39654. [PMID: 18317516 PMCID: PMC2254525 DOI: 10.1155/2007/39654] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2007] [Revised: 07/23/2007] [Accepted: 09/14/2007] [Indexed: 12/21/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are key regulators in various age-associated pathophysiological processes related to energy metabolism and oxidative stress. A progressive rise of oxidative stress and related inflammatory reaction appears the hallmarks of the aging process and many age-related diseases. PPARs are important redox-sensitive
transcription factors and their dyregulated activations seem to be major culprits for these
pathological processes. Drugs targeting PPARs activity are already in widespread clinical use;
however, based on these concepts, this review highlights the understanding of the role of
PPARs in aging and indicates the necessary particular attention for the potential therapeutic
uses of current PPAR agonists in age-associated diseases.
Collapse
|
13
|
Xie Y, Awonuga AO, Zhou S, Puscheck EE, Rappolee DA. Interpreting the stress response of early mammalian embryos and their stem cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2011; 287:43-95. [PMID: 21414586 DOI: 10.1016/b978-0-12-386043-9.00002-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This review analyzes and interprets the normal, pathogenic, and pathophysiological roles of stress and stress enzymes in mammalian development. Emerging data suggest that stem cells from early embryos are induced by stress to perform stress-enzyme-mediated responses that use the strategies of compensatory, prioritized, and reversible differentiation. These strategies have been optimized during evolution and in turn have aspects of energy conservation during stress that optimize and maximize the efficacy of the stress response. It is likely that different types of stem cells have varying degrees of flexibility in mediating compensatory and prioritized differentiation. The significance of this analysis and interpretation is that it will serve as a foundation for yielding tools for diagnosing, understanding normal and pathophysiological mechanisms, and providing methods for managing stress enzymes to improve short- and long-term reproductive outcomes.
Collapse
Affiliation(s)
- Y Xie
- CS Mott Center for Human Growth and Development, Wayne State University School of Medicine, Detroit, Michigan, USA
| | | | | | | | | |
Collapse
|
14
|
Fakhrudin N, Ladurner A, Atanasov AG, Heiss EH, Baumgartner L, Markt P, Schuster D, Ellmerer EP, Wolber G, Rollinger JM, Stuppner H, Dirsch VM. Computer-aided discovery, validation, and mechanistic characterization of novel neolignan activators of peroxisome proliferator-activated receptor gamma. Mol Pharmacol 2010; 77:559-66. [PMID: 20064974 DOI: 10.1124/mol.109.062141] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPAR gamma) agonists are used for the treatment of type 2 diabetes and metabolic syndrome. However, the currently used PPAR gamma agonists display serious side effects, which has led to a great interest in the discovery of novel ligands with favorable properties. The aim of our study was to identify new PPARgamma agonists by a PPAR gamma pharmacophore-based virtual screening of 3D natural product libraries. This in silico approach led to the identification of several neolignans predicted to bind the receptor ligand binding domain (LBD). To confirm this prediction, the neolignans dieugenol, tetrahydrodieugenol, and magnolol were isolated from the respective natural source or synthesized and subsequently tested for PPAR gamma receptor binding. The neolignans bound to the PPAR gamma LBD with EC(50) values in the nanomolar range, exhibiting a binding pattern highly similar to the clinically used agonist pioglitazone. In intact cells, dieugenol and tetrahydrodieugenol selectively activated human PPAR gamma-mediated, but not human PPAR alpha- or -beta/delta-mediated luciferase reporter expression, with a pattern suggesting partial PPAR gamma agonism. The coactivator recruitment study also demonstrated partial agonism of the tested neolignans. Dieugenol, tetrahydrodieugenol, and magnolol but not the structurally related eugenol induced 3T3-L1 preadipocyte differentiation, confirming effectiveness in a cell model with endogenous PPAR gamma expression. In conclusion, we identified neolignans as novel ligands for PPAR gamma, which exhibited interesting activation profiles, recommending them as potential pharmaceutical leads or dietary supplements.
Collapse
Affiliation(s)
- Nanang Fakhrudin
- University of Vienna, Department of Pharmacognosy, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Tzika AA, Mintzopoulos D, Mindrinos M, Zhang J, Rahme LG, Tompkins RG. Microarray analysis suggests that burn injury results in mitochondrial dysfunction in human skeletal muscle. Int J Mol Med 2009; 24:387-92. [PMID: 19639232 DOI: 10.3892/ijmm_00000244] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Burn injuries to extensive areas of the body are complicated by muscle catabolism. Elucidating the molecular mechanisms that mediate this catabolism may facilitate the development of a medical intervention. Here, we assessed the functional classification of genes that were differentially expressed in skeletal muscle following burn injury in 19 children (5.2+/-4.0 years of age), (64+/-15% total burn surface area, TBSA) relative to 13 healthy controls (11.9+/-6.0 years of age). Microarray analysis of samples taken within 10 days of burn injury revealed altered expression of a variety of genes, including some involved in cell and organelle organization and biogenesis, stress response, wound response, external stimulus response, regulation of apoptosis and intracellular signaling. The genes that encode peroxisome proliferator-activated receptors (PPARs; 3 isotypes PPARalpha, PPARgamma and PPARdelta also known as PPARbeta or PPARbeta/delta), which may serve as transcriptional nodal points and therapeutic targets for metabolic syndromes, were among those affected. In particular, expression of the main mitochondrial biogenesis factor PPARgamma-1beta (or PGC-1beta) was downregulated (P<0.0001), while the expression of PPARdelta was upregulated (P<0.001). Expression of PGC-1alpha, the closest homolog of PGC-1beta was upregulated (P=0.0037), and expression of the gene encoding mitochodrial uncoupling protein 2 (UCP2) was also upregulated (P=0.008). These results suggest that altered PPAR and mitochondrial gene expression soon after burn injury may lead to metabolic and mitochondrial dysfunction in human skeletal muscle.
Collapse
Affiliation(s)
- A Aria Tzika
- NMR Surgical Laboratory, Department of Surgery, Massachusetts General and Shriners Hospitals, Harvard Medical School, Boston, MA 02114, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Min Q, Bai YT, Jia G, Wu J, Xiang JZ. High glucose enhances angiotensin-II-mediated peroxisome proliferation-activated receptor-gamma inactivation in human coronary artery endothelial cells. Exp Mol Pathol 2009; 88:133-7. [PMID: 19796634 DOI: 10.1016/j.yexmp.2009.09.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2009] [Accepted: 09/21/2009] [Indexed: 01/11/2023]
Abstract
Activation of the renin-angiotensin system plays an important role in the pathogenesis of vascular complications of hyperglycemia. Clinical studies have demonstrated that hypoglycemic effects of peroxisome proliferation-activated receptor-gamma (PPAR-gamma) activation is potentially associated with a significant decrease of cardiovascular disease events in diabetes patients. We assessed the effect of high glucose on the angiotensin II (Ang II), which induced the inactivation of PPAR-gamma and its signal pathways in human coronary artery endothelial cells (HCAECs). The expression of angiotensin II receptor I (AT1R) protein was analyzed by Western blot and knocked down using siRNA. PPAR-gamma activation was examined using a luminometer and a Dual Luciferase Reporter Assay System. Adhesion molecule expressions of HCAECs were measured using ELISA. Both high glucose and Ang II induced a progressive increase in AT1R protein expression on the HCAECs. Troglitazone, a PPAR-gamma activator, significantly increased the transcription activity of PPAR-gamma in HCAECs in vitro. However, activation of PPAR-gamma was significantly inhibited by high glucose and Ang II stimulation. Furthermore, silencing of AT1R expression was able to inhibit the inactivation of PPAR-gamma induced by Ang II and high glucose. Meanwhile, expression of proinflammatory adhesion molecules was increased by high glucose and Ang II in HCAECs, which is blocked by troglitazone and silencing of AT1R expression. These data strongly suggest high glucose enhanced Ang-II-mediated peroxisome proliferation-activated receptor-gamma inactivation and expression of proinflammatory adhesion molecules in human coronary artery endothelial cells.
Collapse
MESH Headings
- Angiotensin II/genetics
- Angiotensin II/metabolism
- Angiotensin II/pharmacology
- Cell Adhesion Molecules/drug effects
- Cell Adhesion Molecules/metabolism
- Cells, Cultured
- Chromans/pharmacology
- Coronary Vessels/drug effects
- Coronary Vessels/metabolism
- Coronary Vessels/pathology
- Dose-Response Relationship, Drug
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Gene Silencing
- Glucose/pharmacology
- Humans
- PPAR gamma/drug effects
- PPAR gamma/metabolism
- RNA, Small Interfering/genetics
- Receptor, Angiotensin, Type 1/genetics
- Receptor, Angiotensin, Type 1/metabolism
- Thiazolidinediones/pharmacology
- Troglitazone
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Qing Min
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, China.
| | | | | | | | | |
Collapse
|
17
|
Isabel Panadero M, González MDC, Herrera E, Bocos C. Modulación del PPARα por agentes farmacológicos y naturales y sus implicaciones metabólicas. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2008. [DOI: 10.1016/s0214-9168(08)75789-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|