101
|
Brust R, Lin H, Fuhrmann J, Asteian A, Kamenecka TM, Kojetin DJ. Modification of the Orthosteric PPARγ Covalent Antagonist Scaffold Yields an Improved Dual-Site Allosteric Inhibitor. ACS Chem Biol 2017; 12:969-978. [PMID: 28165718 DOI: 10.1021/acschembio.6b01015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
GW9662 and T0070907 are widely used commercially available irreversible antagonists of peroxisome proliferator-activated receptor gamma (PPARγ). These antagonists covalently modify Cys285 located in an orthosteric ligand-binding pocket embedded in the PPARγ ligand-binding domain and are used to block binding of other ligands. However, we recently identified an alternate/allosteric ligand-binding site in the PPARγ LBD to which ligand binding is not inhibited by these orthosteric covalent antagonists. Here, we developed a series of analogs based on the orthosteric covalent antagonist scaffold with the goal of inhibiting both orthosteric and allosteric cellular activation of PPARγ by MRL20, an orthosteric agonist that also binds to an allosteric site. Our efforts resulted in the identification of SR16832 (compound 22), which functions as a dual-site covalent inhibitor of PPARγ transcription by PPARγ-binding ligands. Molecular modeling, protein NMR spectroscopy structural analysis, and biochemical assays indicate the inhibition of allosteric activation occurs in part through expansion of the 2-chloro-5-nitrobenzamidyl orthosteric covalent antagonist toward the allosteric site, weakening of allosteric ligand binding affinity, and inducing conformational changes not competent for cellular PPARγ activation. Furthermore, SR16832 better inhibits binding of rosiglitazone, a thiazolidinedione (TZD) that weakly activates PPARγ when cotreated with orthosteric covalent antagonists, and may better inhibit binding of endogenous PPARγ ligands such as docosahexaenoic acid (DHA) compared to orthosteric covalent antagonists. Compounds such as SR16832 may be useful chemical tools to use as a dual-site bitopic orthosteric and allosteric covalent inhibitor of ligand binding to PPARγ.
Collapse
Affiliation(s)
- Richard Brust
- Department of Molecular Therapeutics,
The Scripps Research Institute, Scripps Florida, Jupiter, Florida 33458, United States
| | - Hua Lin
- Department of Molecular Therapeutics,
The Scripps Research Institute, Scripps Florida, Jupiter, Florida 33458, United States
| | - Jakob Fuhrmann
- Department of Molecular Therapeutics,
The Scripps Research Institute, Scripps Florida, Jupiter, Florida 33458, United States
| | - Alice Asteian
- Department of Molecular Therapeutics,
The Scripps Research Institute, Scripps Florida, Jupiter, Florida 33458, United States
| | - Theodore M. Kamenecka
- Department of Molecular Therapeutics,
The Scripps Research Institute, Scripps Florida, Jupiter, Florida 33458, United States
| | - Douglas J. Kojetin
- Department of Molecular Therapeutics,
The Scripps Research Institute, Scripps Florida, Jupiter, Florida 33458, United States
| |
Collapse
|
102
|
Abstract
Nuclear receptors are a family of transcription factors that can be activated by lipophilic ligands. They are fundamental regulators of development, reproduction, and energy metabolism. In bone, nuclear receptors enable bone cells, including osteoblasts, osteoclasts, and osteocytes, to sense their dynamic microenvironment and maintain normal bone development and remodeling. Our views of the molecular mechanisms in this process have advanced greatly in the past decade. Drugs targeting nuclear receptors are widely used in the clinic for treating patients with bone disorders such as osteoporosis by modulating bone formation and resorption rates. Deficiency in the natural ligands of certain nuclear receptors can cause bone loss; for example, estrogen loss in postmenopausal women leads to osteoporosis and increases bone fracture risk. In contrast, excessive ligands of other nuclear receptors, such as glucocorticoids, can also be detrimental to bone health. Nonetheless, the ligand-induced osteoprotective effects of many other nuclear receptors, e.g., vitamin D receptor, are still in debate and require further characterizations. This review summarizes previous studies on the roles of nuclear receptors in bone homeostasis and incorporates the most recent findings. The advancement of our understanding in this field will help researchers improve the applications of agonists, antagonists, and selective modulators of nuclear receptors for therapeutic purposes; in particular, determining optimal pharmacological drug doses, preventing side effects, and designing new drugs that are more potent and specific.
Collapse
|
103
|
Zolezzi JM, Santos MJ, Bastías-Candia S, Pinto C, Godoy JA, Inestrosa NC. PPARs in the central nervous system: roles in neurodegeneration and neuroinflammation. Biol Rev Camb Philos Soc 2017; 92:2046-2069. [PMID: 28220655 DOI: 10.1111/brv.12320] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 12/21/2016] [Accepted: 01/11/2017] [Indexed: 12/20/2022]
Abstract
Over 25 years have passed since peroxisome proliferators-activated receptors (PPARs), were first described. Like other members of the nuclear receptors superfamily, PPARs have been defined as critical sensors and master regulators of cellular metabolism. Recognized as ligand-activated transcription factors, they are involved in lipid, glucose and amino acid metabolism, taking part in different cellular processes, including cellular differentiation and apoptosis, inflammatory modulation and attenuation of acute and chronic neurological damage in vivo and in vitro. Interestingly, PPAR activation can simultaneously reprogram the immune response, stimulate metabolic and mitochondrial functions, promote axonal growth, induce progenitor cells to differentiate into myelinating oligodendrocytes, and improve brain clearance of toxic molecules such as β-amyloid peptide. Although the molecular mechanisms and cross-talk with different molecular pathways are still the focus of intense research, PPARs are considered potential therapeutic targets for several neuropathological conditions, including degenerative disorders such as Alzheimer's, Parkinson's and Huntington's disease. This review considers recent advances regarding PPARs, as well as new PPAR agonists. We focus on the mechanisms behind the neuroprotective effects exerted by PPARs and summarise the roles of PPARs in different pathologies of the central nervous system, especially those associated with degenerative and inflammatory mechanisms.
Collapse
Affiliation(s)
- Juan M Zolezzi
- Centro de Envejecimiento y Regeneración (CARE-UC), P. Catholic University of Chile, PO Box 114-D, 8331150, Santiago, Chile
| | - Manuel J Santos
- Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Sussy Bastías-Candia
- Facultad de Ciencias, Departamento de Biología, Universidad de Tarapacá, Gral. Velásquez 1775, 1000007, Arica, Chile
| | - Claudio Pinto
- Centro de Envejecimiento y Regeneración (CARE-UC), P. Catholic University of Chile, PO Box 114-D, 8331150, Santiago, Chile
| | - Juan A Godoy
- Centro de Envejecimiento y Regeneración (CARE-UC), P. Catholic University of Chile, PO Box 114-D, 8331150, Santiago, Chile.,Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile
| | - Nibaldo C Inestrosa
- Centro de Envejecimiento y Regeneración (CARE-UC), P. Catholic University of Chile, PO Box 114-D, 8331150, Santiago, Chile.,Facultad de Ciencias Biológicas, Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile, Alameda 340, 8331150, Santiago, Chile.,Faculty of Medicine, Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Avoca Street Randwick NSW 2031, Sydney, Australia.,Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, PO Box 113-D, Avenida Bulnes 01855, 6210427, Punta Arenas, Chile
| |
Collapse
|
104
|
La Paglia L, Listì A, Caruso S, Amodeo V, Passiglia F, Bazan V, Fanale D. Potential Role of ANGPTL4 in the Cross Talk between Metabolism and Cancer through PPAR Signaling Pathway. PPAR Res 2017; 2017:8187235. [PMID: 28182091 PMCID: PMC5274667 DOI: 10.1155/2017/8187235] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023] Open
Abstract
The angiopoietin-like 4 (ANGPTL4) protein belongs to a superfamily of secreted proteins structurally related to factors modulating angiogenesis known as angiopoietins. At first, ANGPTL4 has been identified as an adipokine exclusively involved in lipid metabolism, because of its prevalent expression in liver and adipose tissue. This protein regulates lipid metabolism by inhibiting lipoprotein lipase (LPL) activity and stimulating lipolysis of white adipose tissue (WAT), resulting in increased levels of plasma triglycerides (TG) and fatty acids. Subsequently, ANGPTL4 has been shown to be involved in several nonmetabolic and metabolic conditions, both physiological and pathological, including angiogenesis and vascular permeability, cell differentiation, tumorigenesis, glucose homoeostasis, lipid metabolism, energy homeostasis, wound healing, inflammation, and redox regulation. The transcriptional regulation of ANGPTL4 can be modulated by several transcription factors, including PPARα, PPARβ/δ, PPARγ, and HIF-1α, and nutritional and hormonal conditions. Several studies showed that high levels of ANGPTL4 are associated with poor prognosis in patients with various solid tumors, suggesting an important role in cancer onset and progression, metastasis, and anoikis resistance. Here, we have discussed the potential role of ANGPTL4 in mediating the cross talk between metabolic syndromes, such as diabetes and obesity, and cancer through regulation of its expression by PPARs.
Collapse
Affiliation(s)
- Laura La Paglia
- ICAR-CNR, National Research Council of Italy, 90146 Palermo, Italy
| | - Angela Listì
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Stefano Caruso
- Génomique Fonctionnelle des Tumeurs Solides, INSERM, UMR 1162, 75010 Paris, France
| | - Valeria Amodeo
- Samantha Dickson Brain Cancer Unit, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Francesco Passiglia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Viviana Bazan
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| | - Daniele Fanale
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
105
|
Sarma SM, Khare P, Jagtap S, Singh DP, Baboota RK, Podili K, Boparai RK, Kaur J, Bhutani KK, Bishnoi M, Kondepudi KK. Kodo millet whole grain and bran supplementation prevents high-fat diet induced derangements in a lipid profile, inflammatory status and gut bacteria in mice. Food Funct 2017; 8:1174-1183. [DOI: 10.1039/c6fo01467d] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Protective effect of kodo millet whole grain and bran in diet induced obesity.
Collapse
Affiliation(s)
- Siddhartha M. Sarma
- National Agri-Food Biotechnology Institute
- SAS Nagar
- India
- University Institute of Engineering and Technology
- Panjab University
| | | | - Sneha Jagtap
- National Institute of Pharmaceutical Education and Research (NIPER)
- SAS Nagar
- India
| | | | | | - Koteswaraiah Podili
- Division of Biomedical Sciences
- School of Biosciences and Technology
- VIT University
- Vellore
- India
| | | | - Jaspreet Kaur
- University Institute of Engineering and Technology
- Panjab University
- Chandigarh
- India
| | - Kamlesh K. Bhutani
- National Institute of Pharmaceutical Education and Research (NIPER)
- SAS Nagar
- India
| | | | | |
Collapse
|
106
|
Djouad F, Ipseiz N, Luz-Crawford P, Scholtysek C, Krönke G, Jorgensen C. PPARβ/δ: A master regulator of mesenchymal stem cell functions. Biochimie 2016; 136:55-58. [PMID: 27914902 DOI: 10.1016/j.biochi.2016.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/16/2016] [Accepted: 11/22/2016] [Indexed: 02/06/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) have emerged as key regulators of physiological and immunological processes. Recently, one of their members PPARβ/δ has been identified as major player in the maintenance of bone homeostasis, by promoting Wnt signalling activity in osteoblast and mesenchymal stem cells (MSC). PPARβ/δ not only controls the fate of MSC but also regulates their immunosuppressive properties by directly modulating their NF-κB activity. In this review, we discuss how the regulation of PPARβ/δ provides an innovative strategy for an optimisation of MSC-based therapy.
Collapse
Affiliation(s)
- Farida Djouad
- Inserm U1183, Montpellier, F-34295, France; University of Montpellier, Montpellier, F-34000, France.
| | - Natacha Ipseiz
- Cardiff University, Division of Infection and Immunity, Cardiff, United Kingdom
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Carina Scholtysek
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany; Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Gerhard Krönke
- Department of Internal Medicine 3 and Institute for Clinical Immunology, University Hospital Erlangen, Erlangen, Germany; Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Jorgensen
- Inserm U1183, Montpellier, F-34295, France; University of Montpellier, Montpellier, F-34000, France; Service d'Immuno-Rhumatologie, Hôpital Lapeyronie, Montpellier, F-34295, France
| |
Collapse
|
107
|
Echeverría F, Ortiz M, Valenzuela R, Videla LA. Long-chain polyunsaturated fatty acids regulation of PPARs, signaling: Relationship to tissue development and aging. Prostaglandins Leukot Essent Fatty Acids 2016; 114:28-34. [PMID: 27926461 DOI: 10.1016/j.plefa.2016.10.001] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 10/04/2016] [Accepted: 10/05/2016] [Indexed: 12/31/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that function as ligand-dependent transcription factors that can be activated by different types of fatty acids (FAs). Three isoforms of PPARs have been identify, namely, PPARα, PPARβ/δ, and PPARγ, which are able to bind long-chain polyunsaturated FAs (LCPUFAs), n-3 LCPUFAs being bound with greater affinity to achieve activation. FA binding induces a conformational change of the nuclear receptors, triggering the transcription of specific genes including those encoding for various metabolic and cellular processes such as FA β-oxidation and adipogenesis, thus representing key mediators of lipid homeostasis. In addition, PPARs have important roles during placental, embryonal, and fetal development, and in the regulation of processes related to aging comprising oxidative stress, inflammation, and neuroprotection. The aim of this review was to assess the role of FAs as PPARs ligands, in terms of their main functions associated with FA metabolism and their relevance in the prevention and treatment of related pathologies during human life span.
Collapse
Affiliation(s)
| | - Macarena Ortiz
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Rodrigo Valenzuela
- Nutrition Department, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Luis A Videla
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
108
|
Amber-Vitos O, Chaturvedi N, Nachliel E, Gutman M, Tsfadia Y. The effect of regulating molecules on the structure of the PPAR-RXR complex. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1852-1863. [DOI: 10.1016/j.bbalip.2016.09.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 08/25/2016] [Accepted: 09/03/2016] [Indexed: 11/16/2022]
|
109
|
4-Hydroxyalkenal-activated PPARδ mediates hormetic interactions in diabetes. Biochimie 2016; 136:85-89. [PMID: 27768859 DOI: 10.1016/j.biochi.2016.10.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/12/2016] [Indexed: 12/19/2022]
Abstract
Activated peroxisome proliferator-activated receptor-δ (PPARδ) induces the expression of genes encoding enzymes that metabolize fatty acids and carbohydrate. Attempts to identify cellular activators of PPARδ produced large lists of various fatty acids and their metabolic derivatives; however, there is no consensus on specific and selective binding interactions of natural ligands with PPARδ. Most models on binding interactions within the ligand binding domain (LBD) of PPARδ have been derived from analyses of PPARδ-LBD crystals formed with synthetic low molecular weight ligands. Nonetheless, crystals of the whole receptor with natural ligands or of its heterodimer with its cognate retinoid X receptor (RXR) are not yet available for analysis. We have found that 4-hydroxyalkenals, non-enzymatic peroxidation products of polyunsaturated fatty acids (PUFA), namely, 4-hydroxy-2E,6Z-dodecadienal (4-HDDE) and 4-hydroxy-2E-nonenal (4-HNE), activate PPARδ in vascular endothelial cells and insulin-secreting beta cells, respectively. In both cases activated PPARδ induced adaptive responses that allowed the cells to adjust to ambient stressful metabolic conditions. This review article addresses the interactions of 4-hydroxyalkenals with PPARδ and the resulting hormetic interactions in cells exposed to nutrient overload conditions.
Collapse
|
110
|
Beyaz S, Yilmaz ÖH. Molecular Pathways: Dietary Regulation of Stemness and Tumor Initiation by the PPAR-δ Pathway. Clin Cancer Res 2016; 22:5636-5641. [PMID: 27702819 DOI: 10.1158/1078-0432.ccr-16-0775] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/06/2016] [Accepted: 09/07/2016] [Indexed: 12/30/2022]
Abstract
Peroxisome proliferator-activated receptor delta (PPAR-δ) is a nuclear receptor transcription factor that regulates gene expression during development and disease states, such as cancer. However, the precise role of PPAR-δ during tumorigenesis is not well understood. Recent data suggest that PPAR-δ may have context-specific oncogenic and tumor-suppressive roles depending on the tissue, cell-type, or diet-induced physiology in question. For example, in the intestine, pro-obesity diets, such as a high-fat diet (HFD), are associated with increased colorectal cancer incidence. Interestingly, many of the effects of an HFD in the stem and progenitor cell compartment are driven by a robust PPAR-δ program and contribute to the early steps of intestinal tumorigenesis. Importantly, the PPAR-δ pathway or its downstream mediators may serve as therapeutic intervention points or biomarkers in colon cancer that arise in patients who are obese. Although potent PPAR-δ agonists and antagonists exist, their clinical utility may be enhanced by uncovering how PPAR-δ mediates tumorigenesis in diverse tissues and cell types as well as in response to diet. Clin Cancer Res; 22(23); 5636-41. ©2016 AACR.
Collapse
Affiliation(s)
- Semir Beyaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Division of Hematology/Oncology, Boston Children's Hospital and Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts
| | - Ömer H Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, Massachusetts. .,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts.,Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
111
|
Obermoser V, Urban ME, Murgueitio MS, Wolber G, Kintscher U, Gust R. New telmisartan-derived PPARγ agonists: Impact of the 3D-binding mode on the pharmacological profile. Eur J Med Chem 2016; 124:138-152. [PMID: 27569195 DOI: 10.1016/j.ejmech.2016.08.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/12/2016] [Accepted: 08/13/2016] [Indexed: 11/25/2022]
Abstract
In previous studies, the 4'-((2-propyl-1H-benzo[d]imidazol-1-yl)methyl)-[1,1'-biphenyl]-2-carboxylic acid was identified as pharmacophoric core for PPARγ activation. In this structure-activity relationship study the C2-alkyl chain was elongated and the 2-COOH group was changed to a carbamide/carbonitrile or shifted to the 3- or 4-position. Furthermore, the benzo[d]imidazole was exchanged by 2,3-dihydrobenzo[d]thiazole or 1H-indole. C2-propyl derivatives showed the profile of partial agonists, while elongation of the C2-chain to that of an n-heptyl group or a 4-COOH shift changed the pharmacological profile to that of a potent full agonist. This finding can be explained by binding to the LBD in different ligand conformations. Two anchoring points (Tyr473 and Arg288) exist in the LBD, which have to be contacted to achieve receptor activation. In a crystal violet chemosensitivity assay using COS-7 cells and LNCaP cells expressing PPARγ only the carbamide derivatives influenced the cell growth, independently on the presence of the PPARγ. Therefore, receptor mediated cytotoxicity can be excluded.
Collapse
Affiliation(s)
- Victoria Obermoser
- Pharmaceutical Chemistry, Institute of Pharmacy, Universität Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Margarethe E Urban
- Pharmaceutical Chemistry, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195, Berlin, Germany
| | - Manuela S Murgueitio
- Computer-Aided Drug Design, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195, Berlin, Germany
| | - Gerhard Wolber
- Computer-Aided Drug Design, Institute of Pharmacy, Freie Universität Berlin, Königin-Luise Str. 2+4, 14195, Berlin, Germany
| | - Ulrich Kintscher
- Institute of Pharmacology, Center for Cardiovascular Research, Charité Universitätsmedizin Berlin, Hessische Str. 3-4, 10115, Berlin, Germany
| | - Ronald Gust
- Pharmaceutical Chemistry, Institute of Pharmacy, Universität Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria.
| |
Collapse
|
112
|
Lendvai Á, Deutsch MJ, Plösch T, Ensenauer R. The peroxisome proliferator-activated receptors under epigenetic control in placental metabolism and fetal development. Am J Physiol Endocrinol Metab 2016; 310:E797-810. [PMID: 26860983 DOI: 10.1152/ajpendo.00372.2015] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 02/02/2016] [Indexed: 01/09/2023]
Abstract
The placental metabolism can adapt to the environment throughout pregnancy to both the demands of the fetus and the signals from the mother. Such adaption processes include epigenetic mechanisms, which alter gene expression and may influence the offspring's health. These mechanisms are linked to the diversity of prenatal environmental exposures, including maternal under- or overnutrition or gestational diabetes. The peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that contribute to the developmental plasticity of the placenta by regulating lipid and glucose metabolism pathways, including lipogenesis, steroidogenesis, glucose transporters, and placental signaling pathways, thus representing a link between energy metabolism and reproduction. Among the PPAR isoforms, PPARγ appears to be the main modulator of mammalian placentation. Certain fatty acids and lipid-derived moieties are the natural activating PPAR ligands. By controlling the amounts of maternal nutrients that go across to the fetus, the PPARs play an important regulatory role in placenta metabolism, thereby adapting to the maternal nutritional status. As demonstrated in animal studies, maternal nutrition during gestation can exert long-term influences on the PPAR methylation pattern in offspring organs. This review underlines the current state of knowledge on the relationship between environmental factors and the epigenetic regulation of the PPARs in placenta metabolism and offspring development.
Collapse
Affiliation(s)
- Ágnes Lendvai
- Center for Liver, Digestive, and Metabolic Diseases, Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuel J Deutsch
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Torsten Plösch
- Department of Obstetrics and Gynecology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands;
| | - Regina Ensenauer
- Research Center, Dr. von Hauner Children's Hospital, Ludwig-Maximilians-Universität München, Munich, Germany; Experimental Pediatrics, Department of General Pediatrics, Pediatric Cardiology, and Neonatology, Heinrich-Heine-University Düsseldorf, Dusseldorf, Germany
| |
Collapse
|
113
|
An integrative data mining approach to identifying adverse outcome pathway signatures. Toxicology 2016; 350-352:49-61. [DOI: 10.1016/j.tox.2016.04.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 03/10/2016] [Accepted: 04/18/2016] [Indexed: 01/27/2023]
|
114
|
Polvani S, Tarocchi M, Tempesti S, Bencini L, Galli A. Peroxisome proliferator activated receptors at the crossroad of obesity, diabetes, and pancreatic cancer. World J Gastroenterol 2016; 22:2441-2459. [PMID: 26937133 PMCID: PMC4768191 DOI: 10.3748/wjg.v22.i8.2441] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/17/2015] [Accepted: 01/09/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fourth cause of cancer death with an overall survival of 5% at five years. The development of PDAC is characteristically associated to the accumulation of distinctive genetic mutations and is preceded by the exposure to several risk factors. Epidemiology has demonstrated that PDAC risk factors may be non-modifiable risks (sex, age, presence of genetic mutations, ethnicity) and modifiable and co-morbidity factors related to the specific habits and lifestyle. Recently it has become evident that obesity and diabetes are two important modifiable risk factors for PDAC. Obesity and diabetes are complex systemic and intertwined diseases and, over the years, experimental evidence indicate that insulin-resistance, alteration of adipokines, especially leptin and adiponectin, oxidative stress and inflammation may play a role in PDAC. Peroxisome proliferator activated receptor-γ (PPARγ) is a nuclear receptor transcription factor that is implicated in the regulation of metabolism, differentiation and inflammation. PPARγ is a key regulator of adipocytes differentiation, regulates insulin and adipokines production and secretion, may modulate inflammation, and it is implicated in PDAC. PPARγ agonists are used in the treatment of diabetes and oxidative stress-associated diseases and have been evaluated for the treatment of PDAC. PPARγ is at the cross-road of diabetes, obesity, and PDAC and it is an interesting target to pharmacologically prevent PDAC in obese and diabetic patients.
Collapse
|
115
|
Youssef J, Badr M. Peroxisome Proliferator-Activated Receptors Features, Functions, and Future. NUCLEAR RECEPTOR RESEARCH 2015. [DOI: 10.11131/2015/101188] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
116
|
Peroxisome Proliferator-Activated Receptors and the Heart: Lessons from the Past and Future Directions. PPAR Res 2015; 2015:271983. [PMID: 26587015 PMCID: PMC4637490 DOI: 10.1155/2015/271983] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 10/05/2015] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) belong to the nuclear family of ligand activated transcriptional factors and comprise three different isoforms, PPAR-α, PPAR-β/δ, and PPAR-γ. The main role of PPARs is to regulate the expression of genes involved in lipid and glucose metabolism. Several studies have demonstrated that PPAR agonists improve dyslipidemia and glucose control in animals, supporting their potential as a promising therapeutic option to treat diabetes and dyslipidemia. However, substantial differences exist in the therapeutic or adverse effects of specific drug candidates, and clinical studies have yielded inconsistent data on their cardioprotective effects. This review summarizes the current knowledge regarding the molecular function of PPARs and the mechanisms of the PPAR regulation by posttranslational modification in the heart. We also describe the results and lessons learned from important clinical trials on PPAR agonists and discuss the potential future directions for this class of drugs.
Collapse
|
117
|
Lestari SR, Djati MS, Rudijanto A, Fatchiyah F. PPARγ expression by rambutan peel extract in obesity rat model-induced high-calorie diet. Asian Pac J Trop Biomed 2015. [DOI: 10.1016/j.apjtb.2015.01.030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
118
|
Banerjee R, Bultman SJ, Holley D, Hillhouse C, Bain JR, Newgard CB, Muehlbauer MJ, Willis MS. Non-targeted metabolomics of Brg1/Brm double-mutant cardiomyocytes reveals a novel role for SWI/SNF complexes in metabolic homeostasis. Metabolomics 2015; 11:1287-1301. [PMID: 26392817 PMCID: PMC4574504 DOI: 10.1007/s11306-015-0786-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mammalian SWI/SNF chromatin-remodeling complexes utilize either BRG1 or Brm as alternative catalytic subunits to alter the position of nucleosomes and regulate gene expression. Genetic studies have demonstrated that SWI/SNF complexes are required during cardiac development and also protect against cardiovascular disease and cancer. However, Brm constitutive null mutants do not exhibit a cardiomyocyte phenotype and inducible Brg1 conditional mutations in cardiomyocyte do not demonstrate differences until stressed with transverse aortic constriction, where they exhibit a reduction in cardiac hypertrophy. We recently demonstrated the overlapping functions of Brm and Brg1 in vascular endothelial cells and sought here to test if this overlapping function occurred in cardiomyocytes. Brg1/Brm double mutants died within 21 days of severe cardiac dysfunction associated with glycogen accumulation and mitochondrial defects based on histological and ultrastructural analyses. To determine the underlying defects, we performed nontargeted metabolomics analysis of cardiac tissue by GC/MS from a line of Brg1/Brm double-mutant mice, which lack both Brg1 and Brm in cardiomyocytes in an inducible manner, and two groups of controls. Metabolites contributing most significantly to the differences between Brg1/Brm double-mutant and control-group hearts were then determined using the variable importance in projection analysis. Increased cardiac linoleic acid and oleic acid suggest alterations in fatty acid utilization or intake are perturbed in Brg1/Brm double mutants. Conversely, decreased glucose-6-phosphate, fructose-6-phosphate, and myoinositol suggest that glycolysis and glycogen formation are impaired. These novel metabolomics findings provide insight into SWI/SNF-regulated metabolic pathways and will guide mechanistic studies evaluating the role of SWI/SNF complexes in homeostasis and cardiovascular disease prevention.
Collapse
Affiliation(s)
- Ranjan Banerjee
- University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Darcy Holley
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Carolyn Hillhouse
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - James R. Bain
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA. Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Christopher B. Newgard
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA. Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Michael J. Muehlbauer
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Monte S. Willis
- Department of Pathology & Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA. McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
119
|
Schrader M, Costello JL, Godinho LF, Azadi AS, Islinger M. Proliferation and fission of peroxisomes - An update. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1863:971-83. [PMID: 26409486 DOI: 10.1016/j.bbamcr.2015.09.024] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/16/2015] [Accepted: 09/21/2015] [Indexed: 12/23/2022]
Abstract
In mammals, peroxisomes perform crucial functions in cellular metabolism, signalling and viral defense which are essential to the health and viability of the organism. In order to achieve this functional versatility peroxisomes dynamically respond to molecular cues triggered by changes in the cellular environment. Such changes elicit a corresponding response in peroxisomes, which manifests itself as a change in peroxisome number, altered enzyme levels and adaptations to the peroxisomal structure. In mammals the generation of new peroxisomes is a complex process which has clear analogies to mitochondria, with both sharing the same division machinery and undergoing a similar division process. How the regulation of this division process is integrated into the cell's response to different stimuli, the signalling pathways and factors involved, remains somewhat unclear. Here, we discuss the mechanism of peroxisomal fission, the contributions of the various division factors and examine the potential impact of post-translational modifications, such as phosphorylation, on the proliferation process. We also summarize the signalling process and highlight the most recent data linking signalling pathways with peroxisome proliferation.
Collapse
Affiliation(s)
- Michael Schrader
- College of Life and Environmental Sciences, Biosciences, University of Exeter, EX4 4QJ, Exeter Devon, UK; Centre for Cell Biology, Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal.
| | - Joseph L Costello
- College of Life and Environmental Sciences, Biosciences, University of Exeter, EX4 4QJ, Exeter Devon, UK
| | - Luis F Godinho
- Centre for Cell Biology, Department of Biology, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Afsoon S Azadi
- College of Life and Environmental Sciences, Biosciences, University of Exeter, EX4 4QJ, Exeter Devon, UK
| | - Markus Islinger
- Neuroanatomy, Center for Biomedicine and Medical Technology Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| |
Collapse
|
120
|
Activation of peroxisome proliferator-activated receptor gamma is crucial for antitumoral effects of 6-iodolactone. Mol Cancer 2015; 14:168. [PMID: 26376791 PMCID: PMC4573306 DOI: 10.1186/s12943-015-0436-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 08/24/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Molecular iodine (I2) exhibits antiproliferative and apoptotic effects on in vivo and in vitro cancer models. These effects are thought to be mediated by an iodinated arachidonic acid derivative, 6-iodolactone (6IL), and one of the proposed mechanisms is that 6IL activates Peroxisome Proliferator-Activated Receptors type gamma (PPARG). These receptors have been implicated in the inhibition of carcinogenic processes, in addition to their classical role in maintaining lipid and glucose homeostasis. The aim of this study was to determine whether PPARG participates in the 6IL antiproliferative and apoptotic effects on the mammary cancer cell line MCF-7. METHODS The 6IL/PPARG complex was inhibited by the PPARG antagonist GW9662, in both an endogenous and overexpressed (adenoviral vector infection) context, and stable PPARG-knockdown MCF-7 cells (RNA interference, confirmed with hydrolysis probes and Western blot), were used to corroborate the PPARG participation. 6IL effects on proliferation (measured by Trypan Blue exclusion) and apoptosis (phosphatidylserine identification by flow cytometer) were evaluated in conditions of chemical inhibition (GW9662) and silencing (RNA interference). A wound-healing assay was conducted on wild-type and stable PPARG-knockdown MCF-7 cells to evaluate the antimigrational effect of 6IL. Caspase-8 activity was evaluated to determine if the extrinsic pathway is involved in the effects of 6IL and I2 treatment. RESULTS Antiproliferative and pro-apoptotic 6IL effects require the activation of PPARG. In addition, wound-healing assays show that 6IL is able to inhibit MCF-7 cell migration and that PPARG plays a role in this phenomenon. Finally, the data exclude the participation of the extrinsic apoptotic pathway in 6IL- and I2-induced apoptosis. CONCLUSIONS These results support the previously proposed mechanism, in which the I2 effects are mediated by 6IL, and they provide further support for the use of I2 as coadjuvant in breast cancer treatment.
Collapse
|
121
|
Larsen MC, Bushkofsky JR, Gorman T, Adhami V, Mukhtar H, Wang S, Reeder SB, Sheibani N, Jefcoate CR. Cytochrome P450 1B1: An unexpected modulator of liver fatty acid homeostasis. Arch Biochem Biophys 2015; 571:21-39. [PMID: 25703193 DOI: 10.1016/j.abb.2015.02.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/23/2015] [Accepted: 02/10/2015] [Indexed: 12/12/2022]
Abstract
Cytochrome P450 1b1 (Cyp1b1) expression is absent in mouse hepatocytes, but present in liver endothelia and activated stellate cells. Increased expression during adipogenesis suggests a role of Cyp1b1 metabolism in fatty acid homeostasis. Wild-type C57BL/6j (WT) and Cyp1b1-null (Cyp1b1-ko) mice were provided low or high fat diets (LFD and HFD, respectively). Cyp1b1-deletion suppressed HFD-induced obesity, improved glucose tolerance and prevented liver steatosis. Suppression of lipid droplets in sinusoidal hepatocytes, concomitant with enhanced glycogen granules, was a consistent feature of Cyp1b1-ko mice. Cyp1b1 deletion altered the in vivo expression of 560 liver genes, including suppression of PPARγ, stearoyl CoA desaturase 1 (Scd1) and many genes stimulated by PPARα, each consistent with this switch in energy storage mechanism. Ligand activation of PPARα in Cyp1b1-ko mice by WY-14643 was, nevertheless, effective. Seventeen gene changes in Cyp1b1-ko mice correspond to mouse transgenic expression that attenuated diet-induced diabetes. The absence of Cyp1b1 in mouse hepatocytes indicates participation in energy homeostasis through extra-hepatocyte signaling. Extensive sexual dimorphism in hepatic gene expression suggests a developmental impact of estrogen metabolism by Cyp1b1. Suppression of Scd1 and increased leptin turnover support enhanced leptin participation from the hypothalamus. Cyp1b1-mediated effects on vascular cells may underlie these changes.
Collapse
Affiliation(s)
- Michele Campaigne Larsen
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Justin R Bushkofsky
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States
| | - Tyler Gorman
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Vaqar Adhami
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, United States
| | - Hasan Mukhtar
- Department of Dermatology, University of Wisconsin, Madison, WI 53706, United States
| | - Suqing Wang
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States
| | - Scott B Reeder
- Department of Radiology, University of Wisconsin, Madison, WI 53706, United States; Department of Medical Physics, University of Wisconsin, Madison, WI 53706, United States; Department of Biomedical Engineering, University of Wisconsin, Madison, WI 53706, United States; Department of Medicine, University of Wisconsin, Madison, WI 53706, United States; Department of Emergency Medicine, University of Wisconsin, Madison, WI 53706, United States
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin, Madison, WI 53706, United States
| | - Colin R Jefcoate
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, WI 53706, United States; Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, WI 53706, United States; Endocrinology and Reproductive Physiology Program, University of Wisconsin, Madison, WI 53706, United States.
| |
Collapse
|
122
|
Gangwal RP, Damre MV, Das NR, Sharma SS, Sangamwar AT. Biological evaluation and structural insights for design of subtype-selective peroxisome proliferator activated receptor-α (PPAR-α) agonists. Bioorg Med Chem Lett 2015; 25:270-5. [DOI: 10.1016/j.bmcl.2014.11.052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Revised: 11/07/2014] [Accepted: 11/20/2014] [Indexed: 11/30/2022]
|
123
|
Beekmann K, Rubió L, de Haan LHJ, Actis-Goretta L, van der Burg B, van Bladeren PJ, Rietjens IMCM. The effect of quercetin and kaempferol aglycones and glucuronides on peroxisome proliferator-activated receptor-gamma (PPAR-γ). Food Funct 2015; 6:1098-107. [DOI: 10.1039/c5fo00076a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Quercetin and kaempferol increase PPAR-γ mediated gene expression without acting as agonistic ligands; glucuronidation reduces their activity in cell-based assays.
Collapse
Affiliation(s)
- Karsten Beekmann
- Division of Toxicology
- Wageningen University
- 6700EA Wageningen
- The Netherlands
| | - Laura Rubió
- Department of Food Technology
- XaRTA-UTPV
- Escola Tècnica Superior d'Enginyeria Agrària
- Universitat de Lleida
- 25198 Lleida
| | | | | | | | - Peter J. van Bladeren
- Division of Toxicology
- Wageningen University
- 6700EA Wageningen
- The Netherlands
- Nestlé Research Center
| | | |
Collapse
|
124
|
Kaupang Å, Hildonen S, Halvorsen TG, Mortén M, Vik A, Hansen TV. Involvement of covalent interactions in the mode of action of PPARβ/δ antagonists. RSC Adv 2015. [DOI: 10.1039/c5ra15707b] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Investigations on the mode of action of several different chemical modulators of the peroxisome proliferator-activated receptor β/δ (PPARβ/δ) have been reported using MS and NMR experiments.
Collapse
Affiliation(s)
- Åsmund Kaupang
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Siri Hildonen
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Trine G. Halvorsen
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Magnus Mortén
- Department of Chemistry
- University of Oslo
- 0315 Oslo
- Norway
| | - Anders Vik
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| | - Trond Vidar Hansen
- Department of Pharmaceutical Chemistry
- School of Pharmacy
- University of Oslo
- 0316 Oslo
- Norway
| |
Collapse
|
125
|
Siena LA, Ortiz JPA, Leblanc O, Pessino S. PnTgs1-like expression during reproductive development supports a role for RNA methyltransferases in the aposporous pathway. BMC PLANT BIOLOGY 2014; 14:297. [PMID: 25404464 PMCID: PMC4243328 DOI: 10.1186/s12870-014-0297-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Accepted: 10/20/2014] [Indexed: 05/19/2023]
Abstract
BACKGROUND In flowering plants, apomixis (asexual reproduction via seeds) is widely believed to result from failure of key regulators of the sexual female reproductive pathway. In the past few years, both differential display and RNA-seq comparative approaches involving reproductive organs of sexual plants and their apomictic counterparts have yielded extensive lists of candidate genes. Nevertheless, only a limited number of these genes have been functionally characterized, with few clues consequently available for understanding the molecular control of apomixis. We have previously identified several cDNA fragments with high similarity to genes involved in RNA biology and with differential amplification between sexual and apomictic Paspalum notatum plants. Here, we report the characterization of one of these candidates, namely, N69 encoding a protein of the S-adenosyl-L-methionine-dependent methyltransferases superfamily. The purpose of this work was to extend the N69 cDNA sequence and to characterize its expression at different developmental stages in both sexual and apomictic individuals. RESULTS Molecular characterization of the N69 cDNA revealed homology with genes encoding proteins similar to yeast and mammalian trimethylguanosine synthase/PRIP-interacting proteins. These proteins play a dual role as ERK2-controlled transcriptional coactivators and mediators of sn(o)RNA and telomerase RNA cap trimethylation, and participate in mammals and yeast development. The N69-extended sequence was consequently renamed PnTgs1-like. Expression of PnTgs1-like during reproductive development was significantly higher in floral organs of sexual genotypes compared with apomicts. This difference was not detected in vegetative tissues. In addition, expression levels in reproductive tissues of several genotypes were negatively correlated with facultative apomixis rates. Moreover, in situ hybridization observations revealed that PnTgs1-like expression is relatively higher in ovules of sexual plants throughout development, from premeiosis to maturity. Tissues where differential expression is detected include nucellar cells, the site of aposporous initials differentiation in apomictic genotypes. CONCLUSIONS Our results indicate that PnTgs1-like (formerly N69) encodes a trimethylguanosine synthase-like protein whose function in mammals and yeast is critical for development, including reproduction. Our findings also suggest a pivotal role for this candidate gene in nucellar cell fate, as its diminished expression is correlated with initiation of the apomictic pathway in plants.
Collapse
Affiliation(s)
- Lorena A Siena
- />Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, (S2125ZAA) Zavalla, Santa Fe, Argentina
| | - Juan Pablo A Ortiz
- />Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, (S2125ZAA) Zavalla, Santa Fe, Argentina
- />Instituto de Botánica del Nordeste -IBONE- (UNNE-CONICET), Facultad de Ciencias Agrarias, Universidad Nacional del Nordeste, Sargento Cabral 2131, 3400 Corrientes, Argentina
| | - Olivier Leblanc
- />Institut de Recherche pour le Développement, ERL 5300 IRD/CNRS, UMR 232 IRD/Université de Montpellier 2, 911 Avenue Agropolis, Montpellier, France
| | - Silvina Pessino
- />Laboratorio de Biología Molecular, Facultad de Ciencias Agrarias, Universidad Nacional de Rosario, Parque Villarino, (S2125ZAA) Zavalla, Santa Fe, Argentina
| |
Collapse
|
126
|
Vedell PT, Townsend RR, You M, Malone JP, Grubbs CJ, Bland KI, Muccio DD, Atigadda VR, Chen Y, Vignola K, Lubet RA. Global molecular changes in rat livers treated with
RXR
agonists: a comparison using transcriptomics and proteomics. Pharmacol Res Perspect 2014. [DOI: 10.1002/prp2.74] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Affiliation(s)
- Peter T. Vedell
- Department of Pharmacology Medical College of Wisconsin Cancer Center Milwaukee Wisconsin 53226
| | - Reid R. Townsend
- Department of Internal Medicine Washington University School of Medicine St. Louis Missouri 63110
| | - Ming You
- Department of Pharmacology Medical College of Wisconsin Cancer Center Milwaukee Wisconsin 53226
| | - James P. Malone
- Department of Internal Medicine Washington University School of Medicine St. Louis Missouri 63110
| | - Clinton J. Grubbs
- Department of Surgery University of Alabama at Birmingham Birmingham Alabama 35294
| | - Kirby I. Bland
- Department of Surgery University of Alabama at Birmingham Birmingham Alabama 35294
| | - Donald D. Muccio
- Department of Chemistry University of Alabama at Birmingham Birmingham Alabama 35294
| | - Venkatram R. Atigadda
- Department of Chemistry University of Alabama at Birmingham Birmingham Alabama 35294
| | - Yang Chen
- Department of Science Development Metabolon Research Triangle Park North Carolina 27709
| | - Katie Vignola
- Department of Science Development Metabolon Research Triangle Park North Carolina 27709
| | - Ronald A. Lubet
- Chemoprevention Agent Development Research Group National Cancer Institute Rockville Maryland 20892
| |
Collapse
|
127
|
Neels JG, Grimaldi PA. Physiological functions of peroxisome proliferator-activated receptor β. Physiol Rev 2014; 94:795-858. [PMID: 24987006 DOI: 10.1152/physrev.00027.2013] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The peroxisome proliferator-activated receptors, PPARα, PPARβ, and PPARγ, are a family of transcription factors activated by a diversity of molecules including fatty acids and fatty acid metabolites. PPARs regulate the transcription of a large variety of genes implicated in metabolism, inflammation, proliferation, and differentiation in different cell types. These transcriptional regulations involve both direct transactivation and interaction with other transcriptional regulatory pathways. The functions of PPARα and PPARγ have been extensively documented mainly because these isoforms are activated by molecules clinically used as hypolipidemic and antidiabetic compounds. The physiological functions of PPARβ remained for a while less investigated, but the finding that specific synthetic agonists exert beneficial actions in obese subjects uplifted the studies aimed to elucidate the roles of this PPAR isoform. Intensive work based on pharmacological and genetic approaches and on the use of both in vitro and in vivo models has considerably improved our knowledge on the physiological roles of PPARβ in various cell types. This review will summarize the accumulated evidence for the implication of PPARβ in the regulation of development, metabolism, and inflammation in several tissues, including skeletal muscle, heart, skin, and intestine. Some of these findings indicate that pharmacological activation of PPARβ could be envisioned as a therapeutic option for the correction of metabolic disorders and a variety of inflammatory conditions. However, other experimental data suggesting that activation of PPARβ could result in serious adverse effects, such as carcinogenesis and psoriasis, raise concerns about the clinical use of potent PPARβ agonists.
Collapse
Affiliation(s)
- Jaap G Neels
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| | - Paul A Grimaldi
- Institut National de la Santé et de la Recherche Médicale U 1065, Mediterranean Center of Molecular Medicine (C3M), Team "Adaptive Responses to Immuno-metabolic Dysregulations," Nice, France; and Faculty of Medicine, University of Nice Sophia-Antipolis, Nice, France
| |
Collapse
|
128
|
Tu Z, Moss-Pierce T, Ford P, Jiang TA. Syzygium aromaticum L. (Clove) Extract Regulates Energy Metabolism in Myocytes. J Med Food 2014; 17:1003-10. [DOI: 10.1089/jmf.2013.0175] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Zheng Tu
- Technical Innovation Center, McCormick and Company, Inc., Hunt Valley, Maryland, USA
| | - Tijuana Moss-Pierce
- Technical Innovation Center, McCormick and Company, Inc., Hunt Valley, Maryland, USA
| | - Paul Ford
- Technical Innovation Center, McCormick and Company, Inc., Hunt Valley, Maryland, USA
| | - T. Alan Jiang
- Technical Innovation Center, McCormick and Company, Inc., Hunt Valley, Maryland, USA
| |
Collapse
|
129
|
Roh JI, Cheong C, Sung YH, Lee J, Oh J, Lee BS, Lee JE, Gho YS, Kim DK, Park CB, Lee JH, Lee JW, Kang SM, Lee HW. Perturbation of NCOA6 leads to dilated cardiomyopathy. Cell Rep 2014; 8:991-8. [PMID: 25131203 DOI: 10.1016/j.celrep.2014.07.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Revised: 05/31/2014] [Accepted: 07/15/2014] [Indexed: 11/29/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is a progressive heart disease characterized by left ventricular dilation and contractile dysfunction. Although many candidate genes have been identified with mouse models, few of them have been shown to be associated with DCM in humans. Germline depletion of Ncoa6, a nuclear hormone receptor coactivator, leads to embryonic lethality and heart defects. However, it is unclear whether Ncoa6 mutations cause heart diseases in adults. Here, we report that two independent mouse models of NCOA6 dysfunction develop severe DCM with impaired mitochondrial function and reduced activity of peroxisome proliferator-activated receptor δ (PPARδ), an NCOA6 target critical for normal heart function. Sequencing of NCOA6-coding regions revealed three independent nonsynonymous mutations present in 5 of 50 (10%) patients with idiopathic DCM (iDCM). These data suggest that malfunction of NCOA6 can cause DCM in humans.
Collapse
Affiliation(s)
- Jae-Il Roh
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, South Korea; Yonsei Laboratory Animal Research Center, Yonsei University, Seoul 120-749, South Korea
| | - Cheolho Cheong
- Laboratory of Cellular Physiology and Immunology, Institut de Recherches Cliniques de Montréal, Montréal QC H2W 1R7, Canada; Department of Microbiology, Infectiology and Immunology, Université de Montréal, Montreal QC H3T 1J4, Canada
| | - Young Hoon Sung
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, South Korea; Yonsei Laboratory Animal Research Center, Yonsei University, Seoul 120-749, South Korea
| | - Jeehyun Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, South Korea; Yonsei Laboratory Animal Research Center, Yonsei University, Seoul 120-749, South Korea
| | - Jaewon Oh
- Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - Beom Seob Lee
- Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 120-752, South Korea
| | - Jong-Eun Lee
- DNA Link, Inc., Songpa-Gu, Seoul 138-736, South Korea
| | - Yong Song Gho
- Department of Life Sciences, Pohang University of Science and Technology, Gyeongbuk 790-784, South Korea
| | - Duk-Kyung Kim
- Cardiac and Vascular Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do 440-746, South Korea
| | - Chan Bae Park
- Department of Physiology, Ajou University School of Medicine, Suwon 443-380, South Korea
| | - Ji Hyun Lee
- Department of Oral Biology, College of Dentistry, Yonsei University, Seoul 120-752, South Korea
| | - Jae Woon Lee
- Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | - Seok-Min Kang
- Cardiology Division, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 120-752, South Korea; Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul 120-752, South Korea.
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, South Korea; Yonsei Laboratory Animal Research Center, Yonsei University, Seoul 120-749, South Korea.
| |
Collapse
|
130
|
Abu-Amero KK, Kondkar AA, Oystreck DT, Khan AO, Bosley TM. Microdeletions involving Chromosomes 12 and 22 Associated with Syndromic Duane Retraction Syndrome. Ophthalmic Genet 2014; 35:162-9. [DOI: 10.3109/13816810.2014.921317] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Khaled K Abu-Amero
- Department of Ophthalmology, College of Medicine, King Saud University , Riyadh , Saudi Arabia
| | | | | | | | | |
Collapse
|
131
|
Justus J, Weigand E. A Moderate Zinc Deficiency Does Not Impair Gene Expression of PPARα, PPARγ, and Mitochondrial Enoyl-CoA Delta Isomerase in the Liver of Growing Rats. Nutr Metab Insights 2014; 7:29-37. [PMID: 24855375 PMCID: PMC4024054 DOI: 10.4137/nmi.s14003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 03/24/2014] [Accepted: 03/24/2014] [Indexed: 11/12/2022] Open
Abstract
The aim of the study was to investigate the impact of a moderate zinc deficiency and a high intake of polyunsaturated fat on the mRNA expression of peroxisome-proliferator-activated receptor alpha (PPARα), peroxisome-proliferator-activated receptor gamma (PPARγ), and mitochondrial Δ3Δ2-enoyl-CoA isomerase (ECI) in the liver. Weanling rats were assigned to five groups (eight animals each) and fed semi-synthetic, low-carbohydrate diets containing 7 or 50 mg Zn/kg (low-Zn (LZ) or high-Zn (HZ)) and 22% cocoa butter (CB) or 22% safflower (SF) oil for four weeks. One group each was fed the LZ-CB, LZ-SF, or HZ-SF diet free choice, and one group each was fed the HZ-CB and HZ-SF diets in restricted amounts according to intake of the respective LZ diets. The LZ diets markedly lowered growth and zinc concentrations in plasma and femur. Hepatic mRNA levels of PPARα, PPARγ, and ECI were not reduced by the moderate zinc deficiency. Overall, ECI-mRNA abundance was marginally higher in the SF-fed than in the CB-fed animals.
Collapse
Affiliation(s)
- Jennifer Justus
- Dussmann Service Deutschland GmbH, Frankfurt am Main, Germany
| | - Edgar Weigand
- Institute of Animal Nutrition and Nutritional Physiology, Justus Liebig University, Giessen, Germany
| |
Collapse
|
132
|
Slot IGM, Schols AMWJ, Vosse BAH, Kelders MCJM, Gosker HR. Hypoxia differentially regulates muscle oxidative fiber type and metabolism in a HIF-1α-dependent manner. Cell Signal 2014; 26:1837-45. [PMID: 24794533 DOI: 10.1016/j.cellsig.2014.04.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/27/2014] [Indexed: 12/12/2022]
Abstract
Loss of skeletal muscle oxidative fiber types and mitochondrial capacity is a hallmark of chronic obstructive pulmonary disease and chronic heart failure. Based on in vivo human and animal studies, tissue hypoxia has been hypothesized as determinant, but the direct effect of hypoxia on muscle oxidative phenotype remains to be established. Hence, we determined the effect of hypoxia on in vitro cultured muscle cells, including gene and protein expression levels of mitochondrial components, myosin isoforms (reflecting slow-oxidative versus fast-glycolytic fibers), and the involvement of the regulatory PPAR/PGC-1α pathway. We found that hypoxia inhibits the PPAR/PGC-1α pathway and the expression of mitochondrial components through HIF-1α. However, in contrast to our hypothesis, hypoxia stimulated the expression of slow-oxidative type I myosin via HIF-1α. Collectively, this study shows that hypoxia differentially regulates contractile and metabolic components of muscle oxidative phenotype in a HIF-1α-dependent manner.
Collapse
Affiliation(s)
- Ilse G M Slot
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, PO Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Annemie M W J Schols
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, PO Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Bettine A H Vosse
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, PO Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Marco C J M Kelders
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, PO Box 5800, 6202 AZ Maastricht, The Netherlands.
| | - Harry R Gosker
- Department of Respiratory Medicine, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Centre+, PO Box 5800, 6202 AZ Maastricht, The Netherlands.
| |
Collapse
|
133
|
Jeong E, Koo JE, Yeon SH, Kwak MK, Hwang DH, Lee JY. PPARδ deficiency disrupts hypoxia-mediated tumorigenic potential of colon cancer cells. Mol Carcinog 2014; 53:926-37. [PMID: 24610641 DOI: 10.1002/mc.22144] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Revised: 01/21/2014] [Accepted: 02/11/2014] [Indexed: 01/03/2023]
Abstract
Peroxisome proliferator-activated receptor (PPAR) δ is highly expressed in colon epithelial cells and closely linked to colon carcinogenesis. However, the role of PPARδ in colon cancer cells in a hypoxic tumor microenvironment is not fully understood. We found that expression of the tumor-promoting cytokines, IL-8 and VEGF, induced by hypoxia (<1% O2) and deferoxamine (a hypoxia mimetic) was significantly attenuated in PPARδ-deficient HCT116 colon cancer cells. Consequently, PPARδ-knockout colon cancer cells exposed to hypoxia and deferoxamine failed to stimulate endothelial cell vascularization and macrophage migration/proliferation, whereas wild-type cells were able to induce angiogenesis and macrophage activation in response to hypoxic stress. Hypoxic stress induced transcriptional activation of PPARδ, but not its protein expression, in HCT116 cells. Exogenous expression of p300 potentiated deferoxamine-induced PPARδ transactivation, while siRNA knockdown of p300 abolished hypoxia- and deferoxamine-induced PPARδ transactivation. PPARδ associated with p300 upon hypoxic stress as demonstrated by coimmunoprecipitation studies. PI3K inhibitors or siRNA knockdown of Akt suppressed the PPARδ transactivation induced by hypoxia and deferoxamine in HCT116 cells, leading to decreased expression of IL-8 and VEGF. Collectively, these results reveal that PPARδ is required for hypoxic stress-mediated cytokine expression in colon cancer cells, resulting in promotion of angiogenesis, macrophage recruitment, and macrophage proliferation in the tumor microenvironment. p300 and the PI3K/Akt pathway play a role in the regulation of PPARδ transactivation induced by hypoxic stress. Our results demonstrate the positive crosstalk between PPARδ in tumor cells and the hypoxic tumor microenvironment and provide potential therapeutic targets for colon cancer.
Collapse
Affiliation(s)
- Eunshil Jeong
- Integrated Research Institute of Pharmaceutical Sciences, College of Pharmacy, The Catholic University of Korea, Bucheon, Korea
| | | | | | | | | | | |
Collapse
|
134
|
Integrated physiology and systems biology of PPARα. Mol Metab 2014; 3:354-71. [PMID: 24944896 PMCID: PMC4060217 DOI: 10.1016/j.molmet.2014.02.002] [Citation(s) in RCA: 438] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 02/20/2014] [Accepted: 02/21/2014] [Indexed: 12/23/2022] Open
Abstract
The Peroxisome Proliferator Activated Receptor alpha (PPARα) is a transcription factor that plays a major role in metabolic regulation. This review addresses the functional role of PPARα in intermediary metabolism and provides a detailed overview of metabolic genes targeted by PPARα, with a focus on liver. A distinction is made between the impact of PPARα on metabolism upon physiological, pharmacological, and nutritional activation. Low and high throughput gene expression analyses have allowed the creation of a comprehensive map illustrating the role of PPARα as master regulator of lipid metabolism via regulation of numerous genes. The map puts PPARα at the center of a regulatory hub impacting fatty acid uptake, fatty acid activation, intracellular fatty acid binding, mitochondrial and peroxisomal fatty acid oxidation, ketogenesis, triglyceride turnover, lipid droplet biology, gluconeogenesis, and bile synthesis/secretion. In addition, PPARα governs the expression of several secreted proteins that exert local and endocrine functions.
Collapse
|
135
|
Synergistic Antiproliferative Effects of Combined γ -Tocotrienol and PPAR γ Antagonist Treatment Are Mediated through PPAR γ -Independent Mechanisms in Breast Cancer Cells. PPAR Res 2014; 2014:439146. [PMID: 24729783 PMCID: PMC3960771 DOI: 10.1155/2014/439146] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2013] [Accepted: 01/23/2014] [Indexed: 01/16/2023] Open
Abstract
Previous findings showed that the anticancer effects of combined γ-tocotrienol and peroxisome proliferator activated receptor γ (PPARγ) antagonist treatment caused a large reduction in PPARγ expression. However, other studies suggest that the antiproliferative effects of γ-tocotrienol and/or PPARγ antagonists are mediated, at least in part, through PPARγ-independent mechanism(s). Studies were conducted to characterize the role of PPARγ in mediating the effects of combined treatment of γ-tocotrienol with PPARγ agonists or antagonists on the growth of PPARγ negative +SA mammary cells and PPARγ-positive and PPARγ-silenced MCF-7 and MDA-MB-231 breast cancer cells. Combined treatment of γ-tocotrienol with PPARγ antagonist decreased, while combined treatment of γ-tocotrienol with PPARγ agonist increased, growth of all cancer cells. However, treatment with high doses of 15d-PGJ2, an endogenous natural ligand for PPARγ, had no effect on cancer cell growth. Western blot and qRT-PCR studies showed that the growth inhibitory effects of combined γ-tocotrienol and PPARγ antagonist treatment decreased cyclooxygenase (COX-2), prostaglandin synthase (PGDS), and prostaglandin D2 (PGD2) synthesis. In conclusion, the anticancer effects of combined γ-tocotrienol and PPARγ antagonists treatment in PPARγ negative/silenced breast cancer cells are mediated through PPARγ-independent mechanisms that are associated with a downregulation in COX-2, PGDS, and PGD2 synthesis.
Collapse
|
136
|
Grygiel-Górniak B. Peroxisome proliferator-activated receptors and their ligands: nutritional and clinical implications--a review. Nutr J 2014; 13:17. [PMID: 24524207 PMCID: PMC3943808 DOI: 10.1186/1475-2891-13-17] [Citation(s) in RCA: 857] [Impact Index Per Article: 77.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 02/07/2014] [Indexed: 02/06/2023] Open
Abstract
Peroxisome proliferator-activated receptors are expressed in many tissues, including adipocytes, hepatocytes, muscles and endothelial cells; however, the affinity depends on the isoform of PPAR, and different distribution and expression profiles, which ultimately lead to different clinical outcomes. Because they play an important role in lipid and glucose homeostasis, they are called lipid and insulin sensors. Their actions are limited to specific tissue types and thus, reveal a characteristic influence on target cells. PPARα mainly influences fatty acid metabolism and its activation lowers lipid levels, while PPARγ is mostly involved in the regulation of the adipogenesis, energy balance, and lipid biosynthesis. PPARβ/δ participates in fatty acid oxidation, mostly in skeletal and cardiac muscles, but it also regulates blood glucose and cholesterol levels. Many natural and synthetic ligands influence the expression of these receptors. Synthetic ligands are widely used in the treatment of dyslipidemia (e.g. fibrates--PPARα activators) or in diabetes mellitus (e.g. thiazolidinediones--PPARγ agonists). New generation drugs--PPARα/γ dual agonists--reveal hypolipemic, hypotensive, antiatherogenic, anti-inflammatory and anticoagulant action while the overexpression of PPARβ/δ prevents the development of obesity and reduces lipid accumulation in cardiac cells, even during a high-fat diet. Precise data on the expression and function of natural PPAR agonists on glucose and lipid metabolism are still missing, mostly because the same ligand influences several receptors and a number of reports have provided conflicting results. To date, we know that PPARs have the capability to accommodate and bind a variety of natural and synthetic lipophilic acids, such as essential fatty acids, eicosanoids, phytanic acid and palmitoylethanolamide. A current understanding of the effects of PPARs, their molecular mechanisms and the role of these receptors in nutrition and therapeutic treatment are delineated in this paper.
Collapse
Affiliation(s)
- Bogna Grygiel-Górniak
- Department of Bromatology and Human Nutrition, University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
137
|
McMullen PD, Bhattacharya S, Woods CG, Sun B, Yarborough K, Ross SM, Miller ME, McBride MT, LeCluyse EL, Clewell RA, Andersen ME. A map of the PPARα transcription regulatory network for primary human hepatocytes. Chem Biol Interact 2014; 209:14-24. [DOI: 10.1016/j.cbi.2013.11.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2013] [Revised: 10/14/2013] [Accepted: 11/13/2013] [Indexed: 02/07/2023]
|
138
|
Mansour M. The Roles of Peroxisome Proliferator-Activated Receptors in the Metabolic Syndrome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:217-66. [DOI: 10.1016/b978-0-12-800101-1.00007-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
139
|
Yu J, Xiao Y, Liu J, Ji Y, Liu H, Xu J, Jin X, Liu L, Guan MX, Jiang P. Loss of MED1 triggers mitochondrial biogenesis in C2C12 cells. Mitochondrion 2013; 14:18-25. [PMID: 24368311 DOI: 10.1016/j.mito.2013.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Revised: 10/31/2013] [Accepted: 12/12/2013] [Indexed: 01/09/2023]
Abstract
Under stress conditions transcription factors, including their coactivators, play major roles in mitochondrial biogenesis and oxidative phosphorylation. MED1 (Mediator complex subunit 1) functions as a coactivator of several transcription factors and is implicated in adipogenesis of the lipid and glucose metabolism. This suggests that MED1 may play a role in mitochondrial function. In this study, we found that both the mtDNA content and mitochondrial mass were markedly increased and cell proliferation markedly suppressed in MED1-deficient cells. Upon MED1 loss, Nrf1 and its downstream target genes involved in mitochondrial biogenesis (Tfam, Plormt, Tfb1m), were up-regulated as were those genes in the OXPHOS pathway. Moreover, the knockdown of MED1 resulted in significant changes in the profile of mitochondrial respiration, accompanied by a prominent decrease in the generation of ATP. Collectively, these observations strongly suggest that MED1 has an important affect on mitochondrial function. This further elucidates the role of MED1, particularly its role in the energy metabolism.
Collapse
Affiliation(s)
- Jialing Yu
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yun Xiao
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junxia Liu
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yanchun Ji
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hao Liu
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jing Xu
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaofen Jin
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Li Liu
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Min-Xin Guan
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Pingping Jiang
- Department of Genetics, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
140
|
Vamecq J, Cherkaoui-Malki M, Andreoletti P, Latruffe N. The human peroxisome in health and disease: the story of an oddity becoming a vital organelle. Biochimie 2013; 98:4-15. [PMID: 24075875 DOI: 10.1016/j.biochi.2013.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 09/18/2013] [Indexed: 12/18/2022]
Abstract
Since the first report by Rhodin in 1954, our knowledge on mammalian microbodies/peroxisomes has known several periods. An initial two decades period (1954-1973) has contributed to the biochemical individualisation of peroxisomes as a new class of subcellular organelles (de Duve, 1965). The corresponding research period failed to define a clear role of mammalian peroxisomes in vital functions and intermediary metabolism, explaining why feeling that peroxisomes might be in the human cell oddities has prevailed during several decades. The period standing from 1973 to nowadays has progressively removed this cell oddity view of peroxisomes by highlighting vital function and metabolic role of peroxisomes in health and disease along with genetic and metabolic regulation of peroxisomal protein content, organelle envelope formation and protein signal targeting mechanisms. Research on peroxisomes and their response to various drugs and metabolites, dietary and physiological conditions has also played a key role in the discovery of peroxisome proliferator activated receptors (PPARs) belonging to the nuclear hormone receptor superfamily and for which impact in science and medicine goes now by far beyond that of the peroxisomes.
Collapse
Affiliation(s)
- Joseph Vamecq
- INSERM, Laboratory of Biochemistry and Molecular Biology, Hormonology-Metabolism-Nutrition-Oncology, Centre of Biology and Pathology (CBP), CHU Lille, France.
| | - Mustapha Cherkaoui-Malki
- Laboratory of Biochemistry of Peroxisome, Inflammation & Lipids Metabolism (BioPeroxIL-EA7270), University of Burgundy, 21000 Dijon, France
| | - Pierre Andreoletti
- Laboratory of Biochemistry of Peroxisome, Inflammation & Lipids Metabolism (BioPeroxIL-EA7270), University of Burgundy, 21000 Dijon, France
| | - Norbert Latruffe
- Laboratory of Biochemistry of Peroxisome, Inflammation & Lipids Metabolism (BioPeroxIL-EA7270), University of Burgundy, 21000 Dijon, France
| |
Collapse
|
141
|
Viswakarma N, Jia Y, Bai L, Gao Q, Lin B, Zhang X, Misra P, Rana A, Jain S, Gonzalez FJ, Zhu YJ, Thimmapaya B, Reddy JK. The Med1 subunit of the mediator complex induces liver cell proliferation and is phosphorylated by AMP kinase. J Biol Chem 2013; 288:27898-911. [PMID: 23943624 DOI: 10.1074/jbc.m113.486696] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Mediator, a large multisubunit protein complex, plays a pivotal role in gene transcription by linking gene-specific transcription factors with the preinitiation complex and RNA polymerase II. In the liver, the key subunit of the Mediator complex, Med1, interacts with several nuclear receptors and transcription factors to direct gene-specific transcription. Conditional knock-out of Med1 in the liver showed that hepatocytes lacking Med1 did not regenerate following either partial hepatectomy or treatment with certain nuclear receptor activators and failed to give rise to tumors when challenged with carcinogens. We now report that the adenovirally driven overexpression of Med1 in mouse liver stimulates hepatocyte DNA synthesis with enhanced expression of DNA replication, cell cycle control, and liver-specific genes, indicating that Med1 alone is necessary and sufficient for liver cell proliferation. Importantly, we demonstrate that AMP-activated protein kinase (AMPK), an important cellular energy sensor, interacts with, and directly phosphorylates, Med1 in vitro at serine 656, serine 756, and serine 796. AMPK also phosphorylates Med1 in vivo in mouse liver and in cultured primary hepatocytes and HEK293 and HeLa cells. In addition, we demonstrate that PPARα activators increase AMPK-mediated Med1 phosphorylation in vivo. Inhibition of AMPK by compound C decreased hepatocyte proliferation induced by Med1 and also by the PPARα activators fenofibrate and Wy-14,643. Co-treatment with compound C attenuated PPARα activator-inducible fatty acid β-oxidation in liver. Our results suggest that Med1 phosphorylation by its association with AMPK regulates liver cell proliferation and fatty acid oxidation, most likely as a downstream effector of PPARα and AMPK.
Collapse
Affiliation(s)
- Navin Viswakarma
- From the Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Godoy P, Hewitt NJ, Albrecht U, Andersen ME, Ansari N, Bhattacharya S, Bode JG, Bolleyn J, Borner C, Böttger J, Braeuning A, Budinsky RA, Burkhardt B, Cameron NR, Camussi G, Cho CS, Choi YJ, Craig Rowlands J, Dahmen U, Damm G, Dirsch O, Donato MT, Dong J, Dooley S, Drasdo D, Eakins R, Ferreira KS, Fonsato V, Fraczek J, Gebhardt R, Gibson A, Glanemann M, Goldring CEP, Gómez-Lechón MJ, Groothuis GMM, Gustavsson L, Guyot C, Hallifax D, Hammad S, Hayward A, Häussinger D, Hellerbrand C, Hewitt P, Hoehme S, Holzhütter HG, Houston JB, Hrach J, Ito K, Jaeschke H, Keitel V, Kelm JM, Kevin Park B, Kordes C, Kullak-Ublick GA, LeCluyse EL, Lu P, Luebke-Wheeler J, Lutz A, Maltman DJ, Matz-Soja M, McMullen P, Merfort I, Messner S, Meyer C, Mwinyi J, Naisbitt DJ, Nussler AK, Olinga P, Pampaloni F, Pi J, Pluta L, Przyborski SA, Ramachandran A, Rogiers V, Rowe C, Schelcher C, Schmich K, Schwarz M, Singh B, Stelzer EHK, Stieger B, Stöber R, Sugiyama Y, Tetta C, Thasler WE, Vanhaecke T, Vinken M, Weiss TS, Widera A, Woods CG, Xu JJ, Yarborough KM, Hengstler JG. Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 2013; 87:1315-530. [PMID: 23974980 PMCID: PMC3753504 DOI: 10.1007/s00204-013-1078-5] [Citation(s) in RCA: 965] [Impact Index Per Article: 80.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022]
Abstract
This review encompasses the most important advances in liver functions and hepatotoxicity and analyzes which mechanisms can be studied in vitro. In a complex architecture of nested, zonated lobules, the liver consists of approximately 80 % hepatocytes and 20 % non-parenchymal cells, the latter being involved in a secondary phase that may dramatically aggravate the initial damage. Hepatotoxicity, as well as hepatic metabolism, is controlled by a set of nuclear receptors (including PXR, CAR, HNF-4α, FXR, LXR, SHP, VDR and PPAR) and signaling pathways. When isolating liver cells, some pathways are activated, e.g., the RAS/MEK/ERK pathway, whereas others are silenced (e.g. HNF-4α), resulting in up- and downregulation of hundreds of genes. An understanding of these changes is crucial for a correct interpretation of in vitro data. The possibilities and limitations of the most useful liver in vitro systems are summarized, including three-dimensional culture techniques, co-cultures with non-parenchymal cells, hepatospheres, precision cut liver slices and the isolated perfused liver. Also discussed is how closely hepatoma, stem cell and iPS cell-derived hepatocyte-like-cells resemble real hepatocytes. Finally, a summary is given of the state of the art of liver in vitro and mathematical modeling systems that are currently used in the pharmaceutical industry with an emphasis on drug metabolism, prediction of clearance, drug interaction, transporter studies and hepatotoxicity. One key message is that despite our enthusiasm for in vitro systems, we must never lose sight of the in vivo situation. Although hepatocytes have been isolated for decades, the hunt for relevant alternative systems has only just begun.
Collapse
Affiliation(s)
- Patricio Godoy
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | | | - Ute Albrecht
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Melvin E. Andersen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Nariman Ansari
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Sudin Bhattacharya
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Johannes Georg Bode
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jennifer Bolleyn
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Jan Böttger
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Albert Braeuning
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Robert A. Budinsky
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Britta Burkhardt
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Neil R. Cameron
- Department of Chemistry, Durham University, Durham, DH1 3LE UK
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - J. Craig Rowlands
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General Visceral, and Vascular Surgery, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Olaf Dirsch
- Institute of Pathology, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - María Teresa Donato
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Jian Dong
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dirk Drasdo
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
- INRIA (French National Institute for Research in Computer Science and Control), Domaine de Voluceau-Rocquencourt, B.P. 105, 78153 Le Chesnay Cedex, France
- UPMC University of Paris 06, CNRS UMR 7598, Laboratoire Jacques-Louis Lions, 4, pl. Jussieu, 75252 Paris cedex 05, France
| | - Rowena Eakins
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Karine Sá Ferreira
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
- GRK 1104 From Cells to Organs, Molecular Mechanisms of Organogenesis, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Valentina Fonsato
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Joanna Fraczek
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Andrew Gibson
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Matthias Glanemann
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Chris E. P. Goldring
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
| | - Geny M. M. Groothuis
- Department of Pharmacy, Pharmacokinetics Toxicology and Targeting, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Lena Gustavsson
- Department of Laboratory Medicine (Malmö), Center for Molecular Pathology, Lund University, Jan Waldenströms gata 59, 205 02 Malmö, Sweden
| | - Christelle Guyot
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - David Hallifax
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | - Seddik Hammad
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Adam Hayward
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Claus Hellerbrand
- Department of Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | - Stefan Hoehme
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
| | - Hermann-Georg Holzhütter
- Institut für Biochemie Abteilung Mathematische Systembiochemie, Universitätsmedizin Berlin (Charité), Charitéplatz 1, 10117 Berlin, Germany
| | - J. Brian Houston
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | | | - Kiyomi Ito
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585 Japan
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Claus Kordes
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Edward L. LeCluyse
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Peng Lu
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | - Anna Lutz
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Daniel J. Maltman
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
| | - Madlen Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Patrick McMullen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | | | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jessica Mwinyi
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Andreas K. Nussler
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Jingbo Pi
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Stefan A. Przyborski
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Vera Rogiers
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Cliff Rowe
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Celine Schelcher
- Department of Surgery, Liver Regeneration, Core Facility, Human in Vitro Models of the Liver, Ludwig Maximilians University of Munich, Munich, Germany
| | - Kathrin Schmich
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Michael Schwarz
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Ernst H. K. Stelzer
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Regina Stöber
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama Biopharmaceutical R&D Center, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Ciro Tetta
- Fresenius Medical Care, Bad Homburg, Germany
| | - Wolfgang E. Thasler
- Department of Surgery, Ludwig-Maximilians-University of Munich Hospital Grosshadern, Munich, Germany
| | - Tamara Vanhaecke
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mathieu Vinken
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas S. Weiss
- Department of Pediatrics and Juvenile Medicine, University of Regensburg Hospital, Regensburg, Germany
| | - Agata Widera
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Courtney G. Woods
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | | | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| |
Collapse
|
143
|
Contreras AV, Torres N, Tovar AR. PPAR-α as a key nutritional and environmental sensor for metabolic adaptation. Adv Nutr 2013; 4:439-52. [PMID: 23858092 PMCID: PMC3941823 DOI: 10.3945/an.113.003798] [Citation(s) in RCA: 171] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are transcription factors that belong to the superfamily of nuclear hormone receptors and regulate the expression of several genes involved in metabolic processes that are potentially linked to the development of some diseases such as hyperlipidemia, diabetes, and obesity. One type of PPAR, PPAR-α, is a transcription factor that regulates the metabolism of lipids, carbohydrates, and amino acids and is activated by ligands such as polyunsaturated fatty acids and drugs used to treat dyslipidemias. There is evidence that genetic variants within the PPARα gene have been associated with a risk of the development of dyslipidemia and cardiovascular disease by influencing fasting and postprandial lipid concentrations; the gene variants have also been associated with an acceleration of the progression of type 2 diabetes. The interactions between genetic PPARα variants and the response to dietary factors will help to identify individuals or populations who can benefit from specific dietary recommendations. Interestingly, certain nutritional conditions, such as the prolonged consumption of a protein-restricted diet, can produce long-lasting effects on PPARα gene expression through modifications in the methylation of a specific locus surrounding the PPARα gene. Thus, this review underlines our current knowledge about the important role of PPAR-α as a mediator of the metabolic response to nutritional and environmental factors.
Collapse
Affiliation(s)
- Alejandra V. Contreras
- Faculty of Medicine, National University Autonomous of Mexico, PhD Program in Biomedical Sciences,National Institute of Genomic Medicine
| | - Nimbe Torres
- Nutrition Physiology Department, National Institute of Medical Sciences and Nutrition Salvador Zubirán, Mexico D.F. Mexico
| | - Armando R. Tovar
- Nutrition Physiology Department, National Institute of Medical Sciences and Nutrition Salvador Zubirán, Mexico D.F. Mexico,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
144
|
Kim TH, Kim MY, Jo SH, Park JM, Ahn YH. Modulation of the transcriptional activity of peroxisome proliferator-activated receptor gamma by protein-protein interactions and post-translational modifications. Yonsei Med J 2013; 54:545-59. [PMID: 23549795 PMCID: PMC3635639 DOI: 10.3349/ymj.2013.54.3.545] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) belongs to a nuclear receptor superfamily; members of which play key roles in the control of body metabolism principally by acting on adipose tissue. Ligands of PPARγ, such as thiazolidinediones, are widely used in the treatment of metabolic syndromes and type 2 diabetes mellitus (T2DM). Although these drugs have potential benefits in the treatment of T2DM, they also cause unwanted side effects. Thus, understanding the molecular mechanisms governing the transcriptional activity of PPARγ is of prime importance in the development of new selective drugs or drugs with fewer side effects. Recent advancements in molecular biology have made it possible to obtain a deeper understanding of the role of PPARγ in body homeostasis. The transcriptional activity of PPARγ is subject to regulation either by interacting proteins or by modification of the protein itself. New interacting partners of PPARγ with new functions are being unveiled. In addition, post-translational modification by various cellular signals contributes to fine-tuning of the transcriptional activities of PPARγ. In this review, we will summarize recent advancements in our understanding of the post-translational modifications of, and proteins interacting with, PPARγ, both of which affect its transcriptional activities in relation to adipogenesis.
Collapse
Affiliation(s)
- Tae-Hyun Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Mi-Young Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Seong-Ho Jo
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Joo-Man Park
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Ho Ahn
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
- Brain Korea 21 Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Korea
- Integrative Genomic Research Center for Metabolic Regulation, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
145
|
Bionaz M, Chen S, Khan MJ, Loor JJ. Functional Role of PPARs in Ruminants: Potential Targets for Fine-Tuning Metabolism during Growth and Lactation. PPAR Res 2013; 2013:684159. [PMID: 23737762 PMCID: PMC3657398 DOI: 10.1155/2013/684159] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2012] [Revised: 03/01/2013] [Accepted: 03/01/2013] [Indexed: 12/31/2022] Open
Abstract
Characterization and biological roles of the peroxisome proliferator-activated receptor (PPAR) isotypes are well known in monogastrics, but not in ruminants. However, a wealth of information has accumulated in little more than a decade on ruminant PPARs including isotype tissue distribution, response to synthetic and natural agonists, gene targets, and factors affecting their expression. Functional characterization demonstrated that, as in monogastrics, the PPAR isotypes control expression of genes involved in lipid metabolism, anti-inflammatory response, development, and growth. Contrary to mouse, however, the PPARγ gene network appears to controls milk fat synthesis in lactating ruminants. As in monogastrics, PPAR isotypes in ruminants are activated by long-chain fatty acids, therefore, making them ideal candidates for fine-tuning metabolism in this species via nutrients. In this regard, using information accumulated in ruminants and monogastrics, we propose a model of PPAR isotype-driven biological functions encompassing key tissues during the peripartal period in dairy cattle.
Collapse
Affiliation(s)
- Massimo Bionaz
- Animal and Rangeland Sciences, Oregon State University, Corvallis, OR 97330, USA
| | - Shuowen Chen
- Animal and Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Muhammad J. Khan
- Animal and Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Juan J. Loor
- Animal and Nutritional Sciences, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
146
|
Tu Z, Moss-Pierce T, Ford P, Jiang TA. Rosemary (Rosmarinus officinalis L.) extract regulates glucose and lipid metabolism by activating AMPK and PPAR pathways in HepG2 cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:2803-2810. [PMID: 23432097 DOI: 10.1021/jf400298c] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
An epidemic of metabolic disorders such as obesity and diabetes is rising dramatically. Using natural products as potential preventive and therapeutic interventions for these disorders has drawn worldwide attention. Rosemary has been shown to lower blood glucose and cholesterol levels and mitigate weight gain in several in vivo studies. However, the mechanisms are essentially unknown. We investigated the effects of rosemary extract on metabolism and demonstrated that rosemary extract significantly increased glucose consumption in HepG2 cells. The phosphorylation of AMP-activated protein kinase (AMPK) and its substrate, acetyl-CoA carboxylase (ACC), was increased by rosemary extract. Rosemary extract also transcriptionally regulated the genes involved in metabolism, including SIRT1, PPARγ coactivator 1α (PGC1α), glucose-6-phosphatase (G6Pase), ACC, and low-density lipoprotein receptor (LDLR). Furthermore, the PPARγ-specific antagonist GW9662 diminished rosemary's effects on glucose consumption. Overall, our study suggested that rosemary potentially increases liver glycolysis and fatty acid oxidation by activating AMPK and PPAR pathways.
Collapse
Affiliation(s)
- Zheng Tu
- Technical Innovation Center, McCormick and Company, Inc. , 204 Wight Avenue, Hunt Valley, Maryland 21031, United States
| | | | | | | |
Collapse
|
147
|
Vedell PT, Lu Y, Grubbs CJ, Yin Y, Jiang H, Bland KI, Muccio DD, Cvetkovic D, You M, Lubet R. Effects on gene expression in rat liver after administration of RXR agonists: UAB30, 4-methyl-UAB30, and Targretin (Bexarotene). Mol Pharmacol 2013; 83:698-708. [PMID: 23292798 PMCID: PMC3583492 DOI: 10.1124/mol.112.082404] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 01/04/2013] [Indexed: 12/11/2022] Open
Abstract
Examination of three retinoid X receptor (RXR) agonists [Targretin (TRG), UAB30, and 4-methyl-UAB30 (4-Me-UAB30)] showed that all inhibited mammary cancer in rodents and two (TRG and 4-Me-UAB30) strikingly increased serum triglyceride levels. Agents were administered in diets to female Sprague-Dawley rats. Liver RNA was isolated and microarrayed on the Affymetrix GeneChip Rat Exon 1.0 ST array. Statistical tests identified genes that exhibited differential expression and fell into groups, or modules, with differential expression among agonists. Genes in specific modules were changed by one, two, or all three agonists. An interactome analysis assessed the effects on genes that heterodimerize with known nuclear receptors. For proliferator-activated receptor α/RXR-activated genes, the strongest response was TRG > 4-Me-UAB30 > UAB30. Many liver X receptor/RXR-related genes (e.g., Scd-1 and Srebf1, which are associated with increased triglycerides) were highly expressed in TRG and 4-Me-UAB30- but not UAB30-treated livers. Minimal expression changes were associated with retinoic acid receptor or vitamin D receptor heterodimers by any of the agonists. UAB30 unexpectedly and uniquely activated genes associated with the aryl hydrocarbon hydroxylase (Ah) receptor (Cyp1a1, Cyp1a2, Cyp1b1, and Nqo1). Based on the Ah receptor activation, UAB30 was tested for its ability to prevent dimethylbenzanthracene (DMBA)-induced mammary cancers, presumably by inhibiting DMBA activation, and was highly effective. Gene expression changes were determined by reverse transcriptase-polymerase chain reaction in rat livers treated with Targretin for 2.3, 7, and 21 days. These showed similar gene expression changes at all three time points, arguing some steady-state effect. Different patterns of gene expression among the agonists provided insight into molecular differences and allowed one to predict certain physiologic consequences of agonist treatment.
Collapse
Affiliation(s)
- Peter T Vedell
- Medical College of Wisconsin, Cancer Center, Department of Pharmacology Toxicology, Milwaukee, Wisconsin, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
148
|
Paterniti I, Impellizzeri D, Crupi R, Morabito R, Campolo M, Esposito E, Cuzzocrea S. Molecular evidence for the involvement of PPAR-δ and PPAR-γ in anti-inflammatory and neuroprotective activities of palmitoylethanolamide after spinal cord trauma. J Neuroinflammation 2013; 10:20. [PMID: 23374874 PMCID: PMC3579707 DOI: 10.1186/1742-2094-10-20] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 12/07/2012] [Indexed: 11/10/2022] Open
Abstract
Background Palmitoylethanolamide (PEA) is an endogenous fatty acid amide displaying anti-inflammatory and analgesic actions. Moreover, several data have suggested that PEA reduced inflammation and tissue injury associated with spinal cord trauma and showed a regulatory role for peroxisome proliferator-activated receptor (PPAR)-α signaling in the neuroprotective effect of PEA. However, several other mechanisms could explain the anti-inflammatory and anti-hyperalgesic effects of PEA, including the activation of PPAR-δ and PPAR-γ. The aim of the present study was to carefully investigate the exact contribution of PPAR-δ and PPAR-γ in addition to PPAR-α, in the protective effect of PEA on secondary inflammatory damage associated with an experimental model of spinal cord injury (SCI). Methods SCI was induced in mice through a spinal cord compression by the application of vascular clips (force of 24 g) to the dura via a four-level T5 to T8 laminectomy, and PEA (10 mg/kg, intraperitoneally, 1 and 6 hours after SCI) was injected into wildtype mice and into mice lacking PPAR-α (PPAR-αKO). To deepen the ability of specific PPAR-δ and PPAR-γ antagonists to reverse the effect of PEA, mice were administered GSK0660 or GW9662, 30 minutes before PEA injection. Results Genetic ablation of PPAR-α in mice exacerbated spinal cord damage, while PEA-induced neuroprotection seemed be abolished in PPARαKO mice. Twenty-four hours after spinal cord damage, immunohistological and biochemical studies were performed on spinal cord tissue. Our results indicate that PPAR-δ and PPAR-γ also mediated the protection induced by PEA. In particular, PEA was less effective in PPAR-αKO, GSK0660-treated or GW9662-pretreated mice, as evaluated by the degree of spinal cord inflammation and tissue injury, neutrophil infiltration, proinflammmatory cytokine, inducible nitric oxide synthase expression and motor function. PEA is also able to restore PPAR-δ and PPAR-γ expression in spinal cord tissue. Conclusion This study indicates that PPAR-δ and PPAR-γ can also contribute to the anti-inflammatory activity of PEA in SCI.
Collapse
Affiliation(s)
- Irene Paterniti
- Department of Biological and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, Messina 31-98166, Italy
| | | | | | | | | | | | | |
Collapse
|
149
|
Farràs M, Valls RM, Fernández-Castillejo S, Giralt M, Solà R, Subirana I, Motilva MJ, Konstantinidou V, Covas MI, Fitó M. Olive oil polyphenols enhance the expression of cholesterol efflux related genes in vivo in humans. A randomized controlled trial. J Nutr Biochem 2013; 24:1334-9. [PMID: 23333095 DOI: 10.1016/j.jnutbio.2012.10.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Revised: 10/19/2012] [Accepted: 10/29/2012] [Indexed: 10/27/2022]
Abstract
Both oleic acid and polyphenols have been shown to increase high-density lipoprotein (HDL) cholesterol and to protect HDL from oxidation, a phenomenon associated with a low cholesterol efflux from cells. Our goal was to determine whether polyphenols from olive oil could exert an in vivo nutrigenomic effect on genes related to cholesterol efflux in humans. In a randomized, controlled, cross-over trial, 13 pre/hypertensive patients were assigned 30 ml of two similar olive oils with high (961 mg/kg) and moderate (289 mg/kg) polyphenol content. We found an increase in ATP binding cassette transporter-A1, scavenger receptor class B type 1, peroxisome proliferator-activated receptor (PPAR)BP, PPARα, PPARγ, PPARδ and CD36 gene expression in white blood cells at postprandial after high polyphenol olive oil when compared with moderate polyphenol olive oil intervention (P<.017), with COX-1 reaching borderline significance (P=.024). Linear regression analyses showed that changes in gene expression were related to a decrease in oxidized low-density lipoproteins and with an increase in oxygen radical absorbance capacity and olive oil polyphenols (P<.05). Our results indicate a significant role of olive oil polyphenols in the up-regulation of genes involved in the cholesterol efflux from cells to HDL in vivo in humans. These results are in agreement with previous ones concerning the fact that benefits associated with polyphenol-rich olive oil consumption on cardiovascular risk could be mediated through an in vivo nutrigenomic effect in humans.
Collapse
Affiliation(s)
- Marta Farràs
- Cardiovascular Risk and Nutrition Research Group, CIBER de Fisiopatología de la Obesidad y la Nutrición, IMIM-Research Institut Hospital del Mar, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Transcription Factors and Gene Expression. Mol Pharmacol 2012. [DOI: 10.1002/9781118451908.ch8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
|