1
|
Du X, Mendez-Lara K, Hu S, Diao R, Bhavimani G, Hernandez R, Glass K, De Arruda Saldanha C, Flannick J, Heinz S, Majithia AR. An Alternatively Translated Isoform of PPARG Suggests AF-1 Domain Inhibition as an Insulin Sensitization Target. Diabetes 2025; 74:651-663. [PMID: 39854214 PMCID: PMC11926277 DOI: 10.2337/db24-0497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 01/21/2025] [Indexed: 01/26/2025]
Abstract
ARTICLE HIGHLIGHTS Genetic screens were performed across PPARG to study how disruptive mutations across the full coding sequence affect function. An alternative translational start site in PPARG generates a truncated isoform, peroxisome proliferator-activated receptor γ (PPARγ) M135, which lacks the N-terminal activation function 1 (AF-1) domain and shows increased agonist-induced transactivation of target genes. In human carriers of rare PPARG variants, AF-1 domain-disrupting genetic variants increase agonist-induced PPARγ activity and decrease metabolic syndrome severity. Targeting the AF-1 domain is a potential therapeutic strategy for insulin sensitization.
Collapse
Affiliation(s)
- Xiaomi Du
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
- Bioinformatics and Systems Biology Graduate Program, University of California San Diego, La Jolla, CA
| | - Karen Mendez-Lara
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Siqi Hu
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Rachel Diao
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Guru Bhavimani
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Ruben Hernandez
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Kimberly Glass
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Camila De Arruda Saldanha
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Jason Flannick
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pediatrics, Boston Children’s Hospital, Boston, MA
| | - Sven Heinz
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| | - Amit R. Majithia
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, CA
| |
Collapse
|
2
|
Bombarda-Rocha V, Silva D, Badr-Eddine A, Nogueira P, Gonçalves J, Fresco P. Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer. Cancers (Basel) 2023; 15:4013. [PMID: 37568828 PMCID: PMC10417315 DOI: 10.3390/cancers15154013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Perilipins (PLINs) are the most abundant proteins in lipid droplets (LD). These LD-associated proteins are responsible for upgrading LD from inert lipid storage structures to fully functional organelles, fundamentally integrated in the lipid metabolism. There are five distinct perilipins (PLIN1-5), each with specific expression patterns and metabolic activation, but all capable of regulating the activity of lipases on LD. This plurality creates a complex orchestrated mechanism that is directly related to the healthy balance between lipogenesis and lipolysis. Given the essential role of PLINs in the modulation of the lipid metabolism, these proteins can become interesting targets for the treatment of lipid-associated diseases. Since reprogrammed lipid metabolism is a recognized cancer hallmark, and obesity is a known risk factor for cancer and other comorbidities, the modulation of PLINs could either improve existing treatments or create new opportunities for the treatment of these diseases. Even though PLINs have not been, so far, directly considered for pharmacological interventions, there are many established drugs that can modulate PLINs activity. Therefore, the aim of this study is to assess the involvement of PLINs in diseases related to lipid metabolism dysregulation and whether PLINs can be viewed as potential therapeutic targets for cancer and obesity.
Collapse
Affiliation(s)
- Victória Bombarda-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Allal Badr-Eddine
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
| | - Patrícia Nogueira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
3
|
Schubert M, Becher S, Wallert M, Maeß MB, Abhari M, Rennert K, Mosig AS, Große S, Heller R, Grün M, Lorkowski S. The Peroxisome Proliferator-Activated Receptor (PPAR)- γ Antagonist 2-Chloro-5-Nitro-N-Phenylbenzamide (GW9662) Triggers Perilipin 2 Expression via PPAR δ and Induces Lipogenesis and Triglyceride Accumulation in Human THP-1 Macrophages. Mol Pharmacol 2020; 97:212-225. [PMID: 31871304 DOI: 10.1124/mol.119.117887] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 12/06/2019] [Indexed: 12/20/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are members of the nuclear hormone receptor family, playing pivotal roles in regulating glucose and lipid metabolism as well as inflammation. While characterizing potential PPARγ ligand activity of natural compounds in macrophages, we investigated their influence on the expression of adipophilin [perilipin 2 (PLIN2)], a well-known PPARγ target. To confirm that a compound regulates PLIN2 expression via PPARγ, we performed experiments using the widely used PPARγ antagonist 2-chloro-5-nitro-N-phenylbenzamide (GW9662). Surprisingly, instead of blocking upregulation of PLIN2 expression in THP-1 macrophages, expression was concentration-dependently induced by GW9662 at concentrations and under conditions commonly used. We found that this unexpected upregulation occurs in many human and murine macrophage cell models and also primary cells. Profiling expression of PPAR target genes showed upregulation of several genes involved in lipid uptake, transport, and storage as well as fatty acid synthesis by GW9662. In line with this and with upregulation of PLIN2 protein, GW9662 elevated lipogenesis and increased triglyceride levels. Finally, we identified PPARδ as a mediator of the substantial unexpected effects of GW9662. Our findings show that: 1) the PPARγ antagonist GW9662 unexpectedly activates PPARδ-mediated signaling in macrophages, 2) GW9662 significantly affects lipid metabolism in macrophages, 3) careful validation of experimental conditions and results is required for experiments involving GW9662, and 4) published studies in a context comparable to this work may have reported erroneous results if PPARγ independence was demonstrated using GW9662 only. In light of our findings, certain existing studies might require reinterpretation regarding the role of PPARγ SIGNIFICANCE STATEMENT: Peroxisome proliferator-activated receptors (PPARs) are targets for the treatment of various diseases, as they are key regulators of inflammation as well as lipid and glucose metabolism. Hence, reliable tools to characterize the molecular effects of PPARs are indispensable. We describe profound and unexpected off-target effects of the PPARγ antagonist 2-chloro-5-nitro-N-phenylbenzamide (GW9662) involving PPARδ and in turn affecting macrophage lipid metabolism. Our results question certain existing studies using GW9662 and make better experimental design of future studies necessary.
Collapse
Affiliation(s)
- Martin Schubert
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Stefanie Becher
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Maria Wallert
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Marten B Maeß
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Masoumeh Abhari
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Knut Rennert
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Alexander S Mosig
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Silke Große
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Regine Heller
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Michael Grün
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| | - Stefan Lorkowski
- Institute of Nutrition, Friedrich Schiller University Jena, Jena, Germany (M.S., S.B., M.W., M.B.M., M.A., M.G., S.L.); Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD), Halle-Jena-Leipzig, Germany (M.S., M.W., M.G., S.L.); Institute of Biochemistry II, Jena University Hospital, Jena, Germany (K.R., A.S.M.); and Institute of Molecular Cell Biology, Center for Molecular Biomedicine (CMB), Jena University Hospital and Friedrich Schiller University Jena, Jena, Germany (S.G., R.H.)
| |
Collapse
|
4
|
ADAM8 in invasive cancers: links to tumor progression, metastasis, and chemoresistance. Clin Sci (Lond) 2019; 133:83-99. [PMID: 30635388 DOI: 10.1042/cs20180906] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 11/27/2018] [Accepted: 12/05/2018] [Indexed: 12/26/2022]
Abstract
Ectodomain shedding of extracellular and membrane proteins is of fundamental importance for cell-cell communication in neoplasias. A Disintegrin And Metalloproteinase (ADAM) proteases constitute a family of multifunctional, membrane-bound proteins with traditional sheddase functions. Their protumorigenic potential has been attributed to both, essential (ADAM10 and ADAM17) and 'dispensable' ADAM proteases (ADAM8, 9, 12, 15, and 19). Of specific interest in this review is the ADAM proteinase ADAM8 that has been identified as a significant player in aggressive malignancies including breast, pancreatic, and brain cancer. High expression levels of ADAM8 are associated with invasiveness and predict a poor patient outcome, indicating a prognostic and diagnostic potential of ADAM8. Current knowledge of substrates and interaction partners gave rise to the hypothesis that ADAM8 dysregulation affects diverse processes in tumor biology, attributable to different functional cores of the multidomain enzyme. Proteolytic degradation of extracellular matrix (ECM) components, cleavage of cell surface proteins, and subsequent release of soluble ectodomains promote cancer progression via induction of angiogenesis and metastasis. Moreover, there is increasing evidence for significance of a non-proteolytic function of ADAM8. With the disintegrin (DIS) domain ADAM8 binds integrins such as β1 integrin, thereby activating integrin signaling pathways. The cytoplasmic domain is critical for that activation and involves focal adhesion kinase (FAK), extracellular regulated kinase (ERK1/2), and protein kinase B (AKT/PKB) signaling, further contributing to cancer progression and mediating chemoresistance against first-line therapies. This review highlights the remarkable effects of ADAM8 in tumor biology, concluding that pharmacological inhibition of ADAM8 represents a promising therapeutic approach not only for monotherapy, but also for combinatorial therapies.
Collapse
|
5
|
Elbere I, Silamikelis I, Ustinova M, Kalnina I, Zaharenko L, Peculis R, Konrade I, Ciuculete DM, Zhukovsky C, Gudra D, Radovica-Spalvina I, Fridmanis D, Pirags V, Schiöth HB, Klovins J. Significantly altered peripheral blood cell DNA methylation profile as a result of immediate effect of metformin use in healthy individuals. Clin Epigenetics 2018; 10:156. [PMID: 30545422 PMCID: PMC6293577 DOI: 10.1186/s13148-018-0593-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/29/2018] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Metformin is a widely prescribed antihyperglycemic agent that has been also associated with multiple therapeutic effects in various diseases, including several types of malignancies. There is growing evidence regarding the contribution of the epigenetic mechanisms in reaching metformin's therapeutic goals; however, the effect of metformin on human cells in vivo is not comprehensively studied. The aim of our study was to examine metformin-induced alterations of DNA methylation profiles in white blood cells of healthy volunteers, employing a longitudinal study design. RESULTS Twelve healthy metformin-naïve individuals where enrolled in the study. Genome-wide DNA methylation pattern was estimated at baseline, 10 h and 7 days after the start of metformin administration. The whole-genome DNA methylation analysis in total revealed 125 differentially methylated CpGs, of which 11 CpGs and their associated genes with the most consistent changes in the DNA methylation profile were selected: POFUT2, CAMKK1, EML3, KIAA1614, UPF1, MUC4, LOC727982, SIX3, ADAM8, SNORD12B, VPS8, and several differentially methylated regions as novel potential epigenetic targets of metformin. The main functions of the majority of top-ranked differentially methylated loci and their representative cell signaling pathways were linked to the well-known metformin therapy targets: regulatory processes of energy homeostasis, inflammatory responses, tumorigenesis, and neurodegenerative diseases. CONCLUSIONS Here we demonstrate for the first time the immediate effect of short-term metformin administration at therapeutic doses on epigenetic regulation in human white blood cells. These findings suggest the DNA methylation process as one of the mechanisms involved in the action of metformin, thereby revealing novel targets and directions of the molecular mechanisms underlying the various beneficial effects of metformin. TRIAL REGISTRATION EU Clinical Trials Register, 2016-001092-74. Registered 23 March 2017, https://www.clinicaltrialsregister.eu/ctr-search/trial/2016-001092-74/LV .
Collapse
Affiliation(s)
- Ilze Elbere
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ivars Silamikelis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Monta Ustinova
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ineta Kalnina
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Linda Zaharenko
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Raitis Peculis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ilze Konrade
- Riga East Clinical University Hospital, 2 Hipokrata Street, Riga, LV-1038, Latvia
| | - Diana Maria Ciuculete
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Christina Zhukovsky
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Dita Gudra
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Ilze Radovica-Spalvina
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Davids Fridmanis
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Valdis Pirags
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia
| | - Helgi B Schiöth
- Department of Neuroscience, Functional Pharmacology, Uppsala University, BMC, Box 593, 751 24, Uppsala, Sweden
| | - Janis Klovins
- Latvian Biomedical Research and Study Centre, Ratsupites Str. 1 k-1, Riga, LV-1067, Latvia.
| |
Collapse
|
6
|
de la Rosa Rodriguez MA, Kersten S. Regulation of lipid droplet-associated proteins by peroxisome proliferator-activated receptors. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1212-1220. [DOI: 10.1016/j.bbalip.2017.07.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/24/2022]
|
7
|
Li SQ, Hu ZH, Zhu S, Wang DM, Han HM, Lu HJ. The Effect of ADAM8 on the Proliferation and Apoptosis of Hepatocytes and Hepatoma Carcinoma Cells. J Biochem Mol Toxicol 2015; 29:440-448. [PMID: 26293243 DOI: 10.1002/jbt.21737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 07/22/2015] [Accepted: 08/02/2015] [Indexed: 12/15/2022]
Abstract
This study was undertaken to evaluate the effect of ADAM8 on the proliferation and apoptosis of hepatocytes and hepatoma carcinoma cells during hepatocellular carcinoma (HCC) progression. The expression of ADAM8 was significantly increased with good correlation of PCNA expression increasing and cells apoptosis decreasing during the progression of HCC in the liver of mice. Proliferation experiment in vitro showed that recombinant ADAM8 could induce the expression of PCNA in L02 cells, but not in HepG2 cells. Apoptosis experiment in vitro showed that recombinant ADAM8 did not induce or inhibit the expression of apoptosis-related factors Bcl2, Bax, and Caspase3 in L02 cells, but significantly induced the expression of Bcl2, inhibited the expression of Bax and Caspase3 in HepG2 cells. In conclusion, our study suggested that ADAM8 could promote the proliferation of normal hepatocytes and render hepatoma carcinoma cells more resistant to apoptosis to play important roles during the progression of HCC. ADAM8; Proliferation; Apoptosis.
Collapse
Affiliation(s)
- San-Qiang Li
- The Molecular Medicine Key Laboratory of liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China
| | - Zhi-Hong Hu
- The Molecular Medicine Key Laboratory of liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China
| | - Sha Zhu
- Department of Microbiology Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 45001, People's Republic of China
| | - Dong-Mei Wang
- The Molecular Medicine Key Laboratory of liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China
| | - Hong-Mei Han
- The Molecular Medicine Key Laboratory of liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China
| | - Hua-Jie Lu
- The Molecular Medicine Key Laboratory of liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, 471003, People's Republic of China
| |
Collapse
|
8
|
Li SQ, Wang DM, Zhu S, Ma Z, Li RF, Xu ZS, Han HM. The important role of ADAM8 in the progression of hepatocellular carcinoma induced by diethylnitrosamine in mice. Hum Exp Toxicol 2015; 34:1053-72. [DOI: 10.1177/0960327114567767] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study focuses on investigating the concrete role of a disintegrin and metalloproteinase 8 (ADAM8) in the progression of hepatocellular carcinoma (HCC). Mice received anti-ADAM8 monoclonal antibody (mAb) of 100 μg/100 μl, 200 μg/100 μl or 300 μg/100 μl, respectively, in phosphate-buffered saline (PBS) or PBS intervention during the progression of HCC induced by diethylnitrosamine. The survival rate, body weight, and relative liver weight were determined in the mice. Serum aspartate aminotransferase (AST), alanine aminotransferase (ALT) and α-fetoprotein (AFP) level, hematoxylin–eosin staining, the expression level of vascular endothelial growth factor A (VEGF-A), proliferating cell nuclear antigen (PCNA), caspase 3 (Casp3), B cell leukemia 2 (Bcl2), B cell leukemia 2-associated X protein (Bax), protein p53 (P53), and ADAM8 were detected in the mice at the end of the 24th week. Our results showed that anti-ADAM8 mAb intervention effectively improved the survival rate, reduced the body weight loss and increased the relative liver weight in mice in a dose-dependent manner ( p < 0.05 or p < 0.01). Anti-ADAM8 mAb intervention also significantly lowered serum AST, ALT, and AFP levels ( p < 0.05 or p < 0.01), slowed the progression of HCC ( p < 0.05 or p < 0.01), induced the expression of Casp3, Bax, and P53 ( p < 0.05 or p < 0.01), and inhibited the expression of VEGF-A, PCNA, and Bcl2 in the liver of mice ( p < 0.05 or p < 0.01) in a dose-dependent manner compared with the mice receiving PBS intervention. Our study suggested that ADAM8 might promote the progression of HCC by regulating the expression of these factors. Anti-ADAM8 mAb intervention might be suitable as a potential method for HCC therapy.
Collapse
Affiliation(s)
- S-Q Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - D-M Wang
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - S Zhu
- Department of Microbiology Immunology, College of Basic Medical Sciences, Zhengzhou University, People’s Republic of China
| | - Z Ma
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - R-F Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - Z-S Xu
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| | - H-M Han
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, Luoyang, People’s Republic of China
| |
Collapse
|
9
|
Varela LM, López S, Ortega-Gómez A, Bermúdez B, Buers I, Robenek H, Muriana FJG, Abia R. Postprandial triglyceride-rich lipoproteins regulate perilipin-2 and perilipin-3 lipid-droplet-associated proteins in macrophages. J Nutr Biochem 2014; 26:327-36. [PMID: 25595097 DOI: 10.1016/j.jnutbio.2014.11.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 11/06/2014] [Accepted: 11/06/2014] [Indexed: 02/08/2023]
Abstract
Lipid accumulation in macrophages contributes to atherosclerosis. Within macrophages, lipids are stored in lipid droplets (LDs); perilipin-2 and perilipin-3 are the main LD-associated proteins. Postprandial triglyceride (TG)-rich lipoproteins induce LD accumulation in macrophages. The role of postprandial lipoproteins in perilipin-2 and perilipin-3 regulation was studied. TG-rich lipoproteins (TRLs) induced the levels of intracellular TGs, LDs and perilipin-2 protein expression in THP-1 macrophages and in Apoe(-/-) mice bone-marrow-derived macrophages with low and high basal levels of TGs. Perilipin-3 was only synthesized in mice macrophages with low basal levels of TGs. The regulation was dependent on the fatty acid composition of the lipoproteins; monounsaturated and polyunsaturated fatty acids (PUFAs) more strongly attenuated these effects compared with saturated fatty acids. In THP-1 macrophages, immunofluorescence microscopy and freeze-fracture immunogold labeling indicated that the lipoproteins translocated perilipin-3 from the cytoplasm to the LD surface; only the lipoproteins that were rich in PUFAs suppressed this effect. Chemical inhibition showed that lipoproteins induced perilipin-2 protein expression through the peroxisome proliferator-activated nuclear receptor (PPAR) PPARα and PPARγ pathways. Overall, our data indicate that postprandial TRLs may be involved in atherosclerotic plaque formation through the regulation of perilipin-2 and perilipin-3 proteins in macrophages. Because the fatty acid composition of the lipoproteins is dependent on the type of fat consumed, the ingestion of olive oil, which is rich in monounsaturated fatty acids, and fish oil, which is rich in omega-3 fatty acids, can be considered a good nutritional strategy to reduce the risk of atherosclerosis by LD-associated proteins decrease.
Collapse
Affiliation(s)
- Lourdes M Varela
- Cellular and Molecular Nutrition, Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Sergio López
- Cellular and Molecular Nutrition, Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Almudena Ortega-Gómez
- Cellular and Molecular Nutrition, Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Beatriz Bermúdez
- Department of Pharmacology, School of Pharmacy, University of Seville, Sevilla, Spain
| | - Insa Buers
- Leibniz Institute for Arteriosclerosis Research, Münster, Germany
| | - Horst Robenek
- Institute for Experimental Musculosceletal Medicine, University of Münster, Albert-Schweitzer-Campus 1, Domagkstr. 3, 48149 Münster, Germany
| | - Francisco J G Muriana
- Cellular and Molecular Nutrition, Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Seville, Spain
| | - Rocío Abia
- Cellular and Molecular Nutrition, Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Seville, Spain.
| |
Collapse
|
10
|
Davies NA, Watkeys L, Butcher L, Potter S, Hughes MG, Moir H, Morris K, Thomas AW, Webb R. The contributions of oxidative stress, oxidised lipoproteins and AMPK towards exercise-associated PPARγ signalling within human monocytic cells. Free Radic Res 2014; 49:45-56. [DOI: 10.3109/10715762.2014.978311] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
11
|
Li SQ, Zhu S, Wan XD, Xu ZS, Ma Z. Neutralization of ADAM8 ameliorates liver injury and accelerates liver repair in carbon tetrachloride-induced acute liver injury. J Toxicol Sci 2014; 39:339-51. [PMID: 24646716 DOI: 10.2131/jts.39.339] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Although some studies have described the function of ADAM8 (a disintegrin and metalloprotease 8) related with rheumatoid arthritis, cancer and asthma, etc., the concrete role of ADAM8 in acute liver injury is still unknown. So mice respectively received anti-ADAM8 monoclonal antibody (mAb) of 100 μg/100 μl, 200 μg/100 μl or 300 μg/100 μl in PBS or PBS pre-injection. Then acute liver injury was induced in the mice by intraperitoneal (i.p.) injection of carbon tetrachloride (CCl₄). Serum AST and ALT level, Haematoxylin-eosin (H&E) staining, the expression level of vascular endothelial growth factor (VEGF), cytochrome P450 1A2 (CYP1A2) and proliferating cell nuclear antigen (PCNA) were detected in the mice after CCl4 administration. Our results showed that anti-ADAM8 mAb pre-injection could effectively lower AST and ALT levels (P < 0.05 or P < 0.01) and reduce liver injury (P < 0.05 or P <0.01), induce the expression of VEGF, CYP1A2 and PCNA (P <0.05 or P < 0.01) in dose-dependent manner compared with the control mice which received PBS pre-injection. In summary, our study suggested that ADAM8 might promote liver injury by inhibiting the proliferation of hepatocytes, angiogenesis and affecting the metabolism function of liver during acute liver injury induced by CCl₄. Anti-ADAM8 mAb injection might be suitable as a potential method for acute liver injury therapy.
Collapse
Affiliation(s)
- San-Qiang Li
- The Molecular Medicine Key Laboratory of Liver Injury and Repair, Medical College, Henan University of Science and Technology, China
| | | | | | | | | |
Collapse
|
12
|
Chen J, Jiang X, Duan Y, Long J, Bartsch JW, Deng L. ADAM8 in asthma. Friend or foe to airway inflammation? Am J Respir Cell Mol Biol 2014; 49:875-84. [PMID: 23837412 DOI: 10.1165/rcmb.2013-0168tr] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Airway inflammation has been suggested as the pathological basis in asthma pathogenesis. Recruitment of leukocytes from the vasculature into airway sites is essential for induction of airway inflammation, a process thought to be mediated by a disintegrin and metalloprotease 8 (ADAM8). However, there is an apparent controversy about whether ADAM8 helps or hampers transmigration of leukocytes through endothelium in airway inflammation of asthma. This review outlines the current contradictory concepts concerning the role of ADAM8 in airway inflammation, particularly focusing on the recruitment of leukocytes during asthma, and attempts to bridge the existing experimental data on the basis of the functional analysis of different domains of ADAM8 and their endogenous processing in vivo. We suggest a possible hypothesis for the specific mechanism by which ADAM8 regulates the transmigration of leukocytes to explain the disparity existing in current studies, and we also raise some questions that require future investigations.
Collapse
Affiliation(s)
- Jun Chen
- 1 Key Lab of Biorheological Science and Technology, Ministry of Education, "National 985 Project" Institute of Biorheology and Gene Regulation, Bioengineering College, Chongqing University, Chongqing, P.R. China
| | | | | | | | | | | |
Collapse
|
13
|
Wang J, Pang T, Hafko R, Benicky J, Sanchez-Lemus E, Saavedra JM. Telmisartan ameliorates glutamate-induced neurotoxicity: roles of AT(1) receptor blockade and PPARγ activation. Neuropharmacology 2013; 79:249-61. [PMID: 24316465 DOI: 10.1016/j.neuropharm.2013.11.022] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Revised: 11/22/2013] [Accepted: 11/26/2013] [Indexed: 01/22/2023]
Abstract
Sartans (Angiotensin II AT(1) Receptor Blockers, ARBs) are powerful neuroprotective agents in vivo and protect against IL-1β neurotoxicity in vitro. The purpose of this research was to determine the extent of sartan neuroprotection against glutamate excitotoxicity, a common cause of neuronal injury and apoptosis. The results show that sartans are neuroprotective, significantly reducing glutamate-induced neuronal injury and apoptosis in cultured rat primary cerebellar granule cells (CGCs). Telmisartan was the most potent sartan studied, with an order of potency telmisartan > candesartan > losartan > valsartan. Mechanisms involved reduction of pro-apoptotic caspase-3 activation, protection of the survival PI3K/Akt/GSK-3β pathway and prevention of glutamate-induced ERK1/2 activation. NMDA receptor stimulation was essential for glutamate-induced cell injury and apoptosis. Participation of AT(1A) receptor was supported by glutamate-induced upregulation of AT(1A) gene expression and AT(1) receptor binding. Conversely, AT(1B) or AT(2) receptors played no role. Glutamate-induced neuronal injury and the neuroprotective effect of telmisartan were decreased, but not abolished, in CGCs obtained from AT(1A) knock-out mice. This indicates that although AT(1) receptors are necessary for glutamate to exert its full neurotoxic potential, part of the neuroprotective effect of telmisartan is independent of AT(1) receptor blockade. PPARγ activation was also involved in the neuroprotective effects of telmisartan, as telmisartan enhanced PPARγ nuclear translocation and the PPARγ antagonist GW9662 partially reversed the neuroprotective effects of telmisartan. The present results substantiate the therapeutic use of sartans, in particular telmisartan, in neurodegenerative diseases and traumatic brain disorders where glutamate neurotoxicity plays a significant role.
Collapse
Affiliation(s)
- Juan Wang
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA.
| | - Tao Pang
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA; New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, PR China
| | - Roman Hafko
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA
| | - Julius Benicky
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA.
| | - Enrique Sanchez-Lemus
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA.
| | - Juan M Saavedra
- Section on Pharmacology, Division of Intramural Research Programs, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, 20892, USA; Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC 20057, USA.
| |
Collapse
|
14
|
Zhang W, Wan M, Ma L, Liu X, He J. Protective effects of ADAM8 against cisplatin-mediated apoptosis in non-small-cell lung cancer. Cell Biol Int 2012; 37:47-53. [PMID: 23319321 DOI: 10.1002/cbin.10011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 09/18/2012] [Indexed: 12/31/2022]
Affiliation(s)
- Weifeng Zhang
- Department of Cardiothoracic Surgery; The First Affiliated Hospital of Guangzhou Medical College, State Key Laboratory of Respiratory Disease; Guangzhou 510120 China
| | - Minghui Wan
- Department of Cardiothoracic Surgery; The First Affiliated Hospital of Guangzhou Medical College, State Key Laboratory of Respiratory Disease; Guangzhou 510120 China
| | - Lunchao Ma
- Department of Cardiothoracic Surgery; The First Affiliated Hospital of Guangzhou Medical College, State Key Laboratory of Respiratory Disease; Guangzhou 510120 China
| | - Xiang Liu
- Department of Cardiothoracic Surgery; The First Affiliated Hospital of Guangzhou Medical College, State Key Laboratory of Respiratory Disease; Guangzhou 510120 China
| | - Jianxing He
- Department of Cardiothoracic Surgery; The First Affiliated Hospital of Guangzhou Medical College, State Key Laboratory of Respiratory Disease; Guangzhou 510120 China
| |
Collapse
|
15
|
Telmisartan ameliorates lipopolysaccharide-induced innate immune response through peroxisome proliferator-activated receptor-γ activation in human monocytes. J Hypertens 2012; 30:87-96. [PMID: 22124178 DOI: 10.1097/hjh.0b013e32834dde5f] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Angiotensin II type 1 receptor (AT1) blockers (ARBs) reduce the bacterial endotoxin lipopolysaccharide (LPS)-induced innate immune response in human circulating monocytes expressing few AT1. To clarify the mechanisms of anti-inflammatory effects of ARBs with different peroxisome proliferator-activated receptor-γ (PPARγ)-activating potencies, we focused our study on telmisartan, an ARB with the highest PPARγ-stimulating activity. METHODS Human circulating monocytes and monocytic THP-1 (human acute monocytic leukemia cell line) cells were exposed to 50 ng/ml LPS with or without pre-incubation with telmisartan. AT1 mRNA and protein expressions were determined by real-time PCR and membrane receptor binding assay, respectively. The expression of pro-inflammatory factors was determined by real-time PCR, western blot analysis and ELISA. PPARγ activation was measured by electrophoretic mobility shift assay and its role was determined by pharmacological inhibition and PPARγ gene silencing. RESULTS In human monocytes, telmisartan significantly attenuated the LPS-induced expression of pro-inflammatory factors, the release of pro-inflammatory cytokines and prostaglandin E2, nuclear factor-κB activation and reactive oxygen species formation. In THP-1 cells, telmisartan significantly reduced LPS-induced tumor necrosis factor-α, inhibitor of κB-α, monocyte chemotactic protein-1 (MCP-1) and lectin-like oxidized low-density lipoprotein receptor-1 gene expression and MCP-1-directed migration. Telmisartan also stimulated the expression of the PPARγ target genes cluster of differentiation 36 and ATP-binding cassette subfamily G member 1 in monocytes. The anti-inflammatory effects of telmisartan were prevented by both PPARγ antagonism and PPARγ gene silencing. Anti-inflammatory effects of ARBs correlated with their PPARγ agonist potency. CONCLUSION Our observations demonstrate that in human monocytes, ARBs inhibit the LPS-induced pro-inflammatory response to a major extent through the PPARγ activation pathway and may be beneficial for the treatment of cardiovascular and metabolic disorders in which inflammation plays a major role.
Collapse
|
16
|
Thomas AW, Davies NA, Moir H, Watkeys L, Ruffino JS, Isa SA, Butcher LR, Hughes MG, Morris K, Webb R. Exercise-associated generation of PPARγ ligands activates PPARγ signaling events and upregulates genes related to lipid metabolism. J Appl Physiol (1985) 2012; 112:806-15. [DOI: 10.1152/japplphysiol.00864.2011] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The aim of the present study was to test the hypotheses that exercise is associated with generation of peroxisome proliferator-activated receptor-γ (PPARγ) ligands in the plasma and that this may activate PPARγ signaling within circulating monocytes, thus providing a mechanism to underpin the exercise-induced antiatherogenic benefits observed in previous studies. A cohort of healthy individuals undertook an 8-wk exercise-training program; samples were obtained before (Pre) and after (Post) standardized submaximal exercise bouts (45 min of cycling at 70% of maximal O2 uptake, determined at baseline) at weeks 0, 4, and 8. Addition of plasma samples to PPARγ response element (PPRE)-luciferase reporter gene assays showed increased PPARγ activity following standardized exercise bouts (Post/Pre = 1.23 ± 0.10 at week 0, P < 0.05), suggesting that PPARγ ligands were generated during exercise. However, increases in PPARγ/PPRE-luciferase activity in response to the same standardized exercise bout were blunted during the training program (Post/Pre = 1.18 ± 0.14 and 1.10 ± 0.10 at weeks 4 and 8, respectively, P > 0.05 for both), suggesting that the relative intensity of the exercise may affect PPARγ ligand generation. In untrained individuals, specific transient increases in monocyte expression of PPARγ-regulated genes were observed within 1.5–3 h of exercise (1.7 ± 0.4, 2.6 ± 0.4, and 1.4 ± 0.1 fold for CD36, liver X receptor-α, and ATP-binding cassette subfamily A member 1, respectively, P < 0.05), with expression returning to basal levels within 24 h. In contrast, by the end of the exercise program, expression at the protein level of PPARγ target genes had undergone sustained increases that were not associated with an individual exercise bout (e.g., week 8 Pre/ week 0 Pre = 2.79 ± 0.61 for CD36, P < 0.05). Exercise is known to upregulate PPARγ-controlled genes to induce beneficial effects in skeletal muscle (e.g., mitochondrial biogenesis and aerobic respiration). We suggest that parallel exercise-induced benefits may occur in monocytes, as monocyte PPARγ activation has been linked to beneficial antidiabetic effects (e.g., exercise-induced upregulation of monocytic PPARγ-controlled genes is associated with reverse cholesterol transport and anti-inflammatory effects). Thus, exercise-triggered monocyte PPARγ activation may constitute an additional rationale for prescribing exercise to type 2 diabetes patients.
Collapse
Affiliation(s)
| | | | - H. Moir
- Cardiff School of Health Sciences and
| | | | | | - S. A. Isa
- Cardiff School of Health Sciences and
| | | | - M. G. Hughes
- Cardiff School of Sport, University of Wales Institute Cardiff, Cardiff, United Kingdom
| | - K. Morris
- Cardiff School of Health Sciences and
| | - R. Webb
- Cardiff School of Health Sciences and
| |
Collapse
|
17
|
Molecular Mechanisms and Genome-Wide Aspects of PPAR Subtype Specific Transactivation. PPAR Res 2010; 2010. [PMID: 20862367 PMCID: PMC2938449 DOI: 10.1155/2010/169506] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 06/27/2010] [Indexed: 12/13/2022] Open
Abstract
The peroxisome proliferator-activated receptors (PPARs) are central regulators of fat metabolism, energy homeostasis, proliferation, and inflammation. The three PPAR subtypes, PPARα, β/δ, and γ activate overlapping but also very different target gene programs. This review summarizes the insights into PPAR subtype-specific transactivation provided by genome-wide studies and discusses the recent advances in the understanding of the molecular mechanisms underlying PPAR subtype specificity with special focus on the regulatory role of AF-1.
Collapse
|
18
|
Tsukahara T, Tsukahara R, Fujiwara Y, Yue J, Cheng Y, Guo H, Bolen A, Zhang C, Balazs L, Re F, Du G, Frohman MA, Baker DL, Parrill AL, Uchiyama A, Kobayashi T, Murakami-Murofushi K, Tigyi G. Phospholipase D2-dependent inhibition of the nuclear hormone receptor PPARgamma by cyclic phosphatidic acid. Mol Cell 2010; 39:421-32. [PMID: 20705243 PMCID: PMC3446787 DOI: 10.1016/j.molcel.2010.07.022] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 03/17/2010] [Accepted: 06/14/2010] [Indexed: 01/16/2023]
Abstract
Cyclic phosphatidic acid (1-acyl-2,3-cyclic-glycerophosphate, CPA), one of nature's simplest phospholipids, is found in cells from slime mold to humans and has a largely unknown function. We find here that CPA is generated in mammalian cells in a stimulus-coupled manner by phospholipase D2 (PLD2) and binds to and inhibits the nuclear hormone receptor PPARgamma with nanomolar affinity and high specificity through stabilizing its interaction with the corepressor SMRT. CPA production inhibits the PPARgamma target-gene transcription that normally drives adipocytic differentiation of 3T3-L1 cells, lipid accumulation in RAW264.7 cells and primary mouse macrophages, and arterial wall remodeling in a rat model in vivo. Inhibition of PLD2 by shRNA, a dominant-negative mutant, or a small molecule inhibitor blocks CPA production and relieves PPARgamma inhibition. We conclude that CPA is a second messenger and a physiological inhibitor of PPARgamma, revealing that PPARgamma is regulated by endogenous agonists as well as by antagonists.
Collapse
Affiliation(s)
- Tamotsu Tsukahara
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Ryoko Tsukahara
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Yuko Fujiwara
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Junming Yue
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Yunhui Cheng
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Huazhang Guo
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Alyssa Bolen
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Chunxiang Zhang
- Department of Medicine, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Louisa Balazs
- Department of Pathology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Fabio Re
- Department of Molecular Sciences, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| | - Guangwei Du
- Center for Developmental Genetics and Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Michael A. Frohman
- Center for Developmental Genetics and Department of Pharmacology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel L. Baker
- Department of Chemistry, University of Memphis, TN 38152, USA
| | - Abby L. Parrill
- Department of Chemistry, University of Memphis, TN 38152, USA
- Computational Research on Materials Institute, University of Memphis, TN 38152, USA
| | - Ayako Uchiyama
- Department of Biology, Ochanomizu University, Tokyo 112-8610, Japan
| | | | | | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, TN, 38163, USA
| |
Collapse
|
19
|
Escribano L, Simón AM, Gimeno E, Cuadrado-Tejedor M, López de Maturana R, García-Osta A, Ricobaraza A, Pérez-Mediavilla A, Del Río J, Frechilla D. Rosiglitazone rescues memory impairment in Alzheimer's transgenic mice: mechanisms involving a reduced amyloid and tau pathology. Neuropsychopharmacology 2010; 35:1593-604. [PMID: 20336061 PMCID: PMC3055461 DOI: 10.1038/npp.2010.32] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Clinical studies suggest that agonists at peroxisome proliferator-activated receptor gamma (PPARgamma) may exert beneficial effects in patients with mild-to-moderate Alzheimer's disease (AD), but the mechanism for the potential therapeutic interest of this class of drugs has not yet been elucidated. Here, in mice overexpressing mutant human amyloid precursor protein, we found that chronic treatment with rosiglitazone, a high-affinity agonist at PPARgamma, facilitated beta-amyloid peptide (Abeta) clearance. Rosiglitazone not only reduced Abeta burden in the brain but, importantly, almost completely removed the abundant amyloid plaques observed in the hippocampus and entorhinal cortex of 13-month-old transgenic mice. In the hippocampus, neuropil threads containing phosphorylated tau, probably corresponding to dystrophic neurites, were also decreased by the drug. Rosiglitazone switched on the activated microglial phenotype, promoting its phagocytic ability, reducing the expression of proinflammatory markers and inducing factors for alternative differentiation. The decreased amyloid pathology may account for the reduction of p-tau-containing neuropil threads and for the rescue of impaired recognition and spatial memory in the transgenic mice. This study provides further insights into the mechanisms for the beneficial effect of rosiglitazone in AD patients.
Collapse
Affiliation(s)
- Luis Escribano
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain
| | - Ana-María Simón
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain
| | - Esther Gimeno
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain
| | - Mar Cuadrado-Tejedor
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain
| | | | - Ana García-Osta
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain
| | - Ana Ricobaraza
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain
| | | | - Joaquín Del Río
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain
| | - Diana Frechilla
- Division of Neurosciences, CIMA, University of Navarra, Pamplona, CIBERNED, Spain,Division of Neurosciences, CIMA, University of Navarra, CIBERNED, Av. Pio XII 55, Pamplona 31008, Spain, Tel: +349 4819 4700, Fax: +349 4819 4715, E-mail:
| |
Collapse
|
20
|
Chen FL, Yang ZH, Wang XC, Liu Y, Yang YH, Li LX, Liang WC, Zhou WB, Hu RM. Adipophilin affects the expression of TNF-alpha, MCP-1, and IL-6 in THP-1 macrophages. Mol Cell Biochem 2010; 337:193-199. [PMID: 19851831 DOI: 10.1007/s11010-009-0299-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2009] [Accepted: 10/08/2009] [Indexed: 01/25/2023]
Abstract
Macrophages accumulated in the arterial intima play an important role in the development of atherosclerosis by producing a large number of proinflammatory cytokines which accelerate the disease. Recent studies show that adipophilin might be involved in inflammatory processes in macrophages. In this study, we observe the effect of adipophilin on proinflammatory cytokine expression and secretion in THP-1 macrophages. SiRNA and adipophilin gene overexpression mediated by an pEGFP-C3 vector were used to observe the effect of adipophilin on proinflammatory cytokines in THP-1 macrophages in vitro. Realtime PCR and enzyme-linked immunosorbent assay (ELISA) were applied to detect the production of tumor necrosis factor alpha (TNF-alpha), monocyte chemoattractant protein 1 (MCP-1), and interleukin-6 (IL-6). It was found that acetylated low-density lipoprotein (AcLDL), pioglitazone [a peroxisome proliferator-activated receptor gamma (PPARgamma) agonist] increased adipophilin expression in macrophages, while glucose had no such affect. It was also shown that adipophilin augments TNF-alpha, MCP-1, and IL-6 expression in AcLDL induced macrophages. Our results suggest that adipophilin augment inflammation in macrophages, which might be one role of adipophilin in atherosclerosis.
Collapse
Affiliation(s)
- F L Chen
- Department of Endocrinology, Huashan Hospital, Institute of Endocrinology and Diabetology at Fudan University, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
ADAM8 is a negative regulator of retinal neovascularization and of the growth of heterotopically injected tumor cells in mice. J Mol Med (Berl) 2010; 88:497-505. [PMID: 20119708 DOI: 10.1007/s00109-010-0591-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 12/11/2009] [Accepted: 12/25/2009] [Indexed: 01/20/2023]
Abstract
ADAM8 is a member of the "a disintegrin and metalloproteinase" (ADAM) family of membrane-anchored metalloproteinases. ADAM8-deficient mice have no evident spontaneous developmental or pathological defects, and little is currently known about the role of ADAM8 in disease. Here, we investigated the contribution of ADAM8 to pathological neovascularization in mice using an oxygen-induced retinopathy (OIR) model and heterotopical injection of tumor cells. We found an increase in retinal re-vascularization but fewer neovascular tufts in the OIR model and increased growth of heterotopically injected tumor cells in Adam8-/- mice compared with wild-type controls. These results suggest that ADAM8 functions to limit both of these processes in wild-type mice. In cell-based assays, overexpression of ADAM8 increased the ectodomain shedding of several co-expressed membrane proteins with roles in angiogenesis (CD31, Tie-2, Flk-1, Flt-1, EphrinB2, EphB4, VE-cadherin, KL-1, E-selectin, and neuregulin-1beta2). Thus, dysregulated expression of ADAM8 in endothelial cells in vivo could potentially increase the processing of these and other substrate proteins. Taken together, our findings suggest that inhibiting ADAM8 could be useful for promoting re-vascularization and thereby preventing formation of neovascular tufts in proliferative retinopathies. On the other hand, blocking ADAM8 could be detrimental in the context of rapidly growing tumors.
Collapse
|
22
|
Abedin SA, Thorne JL, Battaglia S, Maguire O, Hornung LB, Doherty AP, Mills IG, Campbell MJ. Elevated NCOR1 disrupts a network of dietary-sensing nuclear receptors in bladder cancer cells. Carcinogenesis 2009; 30:449-56. [PMID: 19126649 PMCID: PMC2722152 DOI: 10.1093/carcin/bgp005] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2008] [Revised: 12/08/2008] [Accepted: 12/26/2008] [Indexed: 01/06/2023] Open
Abstract
Increasingly invasive bladder cancer cells lines displayed insensitivity toward a panel of dietary-derived ligands for members of the nuclear receptor superfamily. Insensitivity was defined through altered gene regulatory actions and cell proliferation and reflected both reduced receptor expression and elevated nuclear receptor corepressor 1 (NCOR1) expression. Stable overexpression of NCOR1 in sensitive cells (RT4) resulted in a panel of clones that recapitulated the resistant phenotype in terms of gene regulatory actions and proliferative responses toward ligand. Similarly, silencing RNA approaches to NCOR1 in resistant cells (EJ28) enhanced ligand gene regulatory and proliferation responses, including those mediated by peroxisome proliferator-activated receptor (PPAR) gamma and vitamin D receptor (VDR) receptors. Elevated NCOR1 levels generate an epigenetic lesion to target in resistant cells using the histone deacetylase inhibitor vorinostat, in combination with nuclear receptor ligands. Such treatments revealed strong-additive interactions toward the PPARgamma, VDR and Farnesoid X-activated receptors. Genome-wide microarray and microfluidic quantitative real-time, reverse transcription-polymerase chain reaction approaches, following the targeting of NCOR1 activity and expression, revealed the selective capacity of this corepressor to govern common transcriptional events of underlying networks. Combined these findings suggest that NCOR1 is a selective regulator of nuclear receptors, notably PPARgamma and VDR, and contributes to their loss of sensitivity. Combinations of epigenetic therapies that target NCOR1 may prove effective, even when receptor expression is reduced.
Collapse
Affiliation(s)
| | | | | | - Orla Maguire
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Laura B. Hornung
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Alan P. Doherty
- Department of Urology, Queen Elizabeth Hospital, Edgbaston, Birmingham B15 2TH, UK
| | - Ian G. Mills
- Uro-Oncology Research Group, Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Moray J. Campbell
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| |
Collapse
|
23
|
Levula M, Airla N, Oksala N, Hernesniemi JA, Pelto-Huikko M, Salenius JP, Zeitlin R, Järvinen O, Huovila APJ, Nikkari ST, Jaakkola O, Ilveskoski E, Mikkelsson J, Perola M, Laaksonen R, Kytömäki L, Soini JT, Kähönen M, Parkkinen J, Karhunen PJ, Lehtimäki T. ADAM8 and its single nucleotide polymorphism 2662 T/G are associated with advanced atherosclerosis and fatal myocardial infarction: Tampere vascular study. Ann Med 2009; 41:497-507. [PMID: 19575316 DOI: 10.1080/07853890903025945] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
OBJECTIVE Previously, we scanned all 23,000 human genes for differential expression between normal and atherosclerotic tissues and found the involvement of ADAM8. METHODS We investigated the expression of ADAM8 mRNA and protein level in human atherosclerotic tissues and non-atherosclerotic internal thoracic arteries as well as the association of ADAM8 2662 T/G single nucleotide polymorphism (SNP) with the extent of coronary atherosclerosis and with the risk of fatal myocardial infarction. RESULTS ADAM8 mRNA was up-regulated in carotid, aortic, and femoral atherosclerotic plaques (n=24) when compared with non-atherosclerotic arteries. ADAM8 protein expression was increased in advanced atherosclerotic plaques as compared to control vessels wherein it was localized to macrophages and smooth muscle cells The G allele carriers of the ADAM8 2662 T/G SNP had significantly larger areas of fibrotic, calcified, and complicated plaques in coronary arteries (P=0.027, P=0.011, and P=0.011, respectively) and significantly higher occurrence of myocardial infarction (MI) (P=0.004) and fatal pre-hospital MI (P=0.003) than did the TT homozygotes. CONCLUSION ADAM8 is a promising candidate to be involved in atherosclerosis, and its 2662 T/G allelic variant significantly associates with advanced atherosclerotic lesion areas and MI.
Collapse
Affiliation(s)
- Mari Levula
- Laboratory of Atherosclerosis Genetics, Centre for Laboratory Medicine, Tampere University Hospital and Department of Clinical Chemistry, Medical School, University of Tampere, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Endolysosomal phospholipidosis and cytosolic lipid droplet storage and release in macrophages. Biochim Biophys Acta Mol Cell Biol Lipids 2008; 1791:524-39. [PMID: 19146988 DOI: 10.1016/j.bbalip.2008.12.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2008] [Revised: 12/08/2008] [Accepted: 12/11/2008] [Indexed: 12/12/2022]
Abstract
This review summarizes the current knowledge of endolysosomal and cytoplasmic lipid storage in macrophages induced by oxidized LDL (Ox-LDL), enzymatically degraded LDL (E-LDL) and other atherogenic lipoprotein modifications, and their relation to the adapter protein 3 (AP-3) dependent ABCA1 and ABCG1 cellular lipid efflux pathways. We compare endolysosomal lipid storage caused either through drug induced phospholipidosis, inheritable endolysosomal and cytosolic lipid storage disorders and Ox-LDL or E-LDL induced phagosomal uptake and cytosolic lipid droplet storage in macrophages. Ox-LDL is resistant to rapid endolysosomal hydrolysis and is trapped within the endolysosomal compartment generating lamellar bodies which resemble the characteristics of phospholipidosis. Various inherited lysosomal storage diseases including sphingolipidosis, glycosphingolipidosis and cholesterylester storage diseases also present a phospholipidosis phenotype. In contrast E-LDL resembling coreless unesterified cholesterol enriched LDL-particles, with a multilamellar, liposome-like structure, lead to rapid phagosomal degradation and cytosolic lipid droplet accumulation. As a consequence the uptake of E-LDL through type I and type II phagocytosis leads to increased lipid droplet formation and moderate upregulation of ABCA1 and ABCG1 while uptake of Ox-LDL leads to a rapid expansion of the lysosomal compartment and a pronounced upregulation of the ABCA1/ABCG1/AP-3 lipid efflux pathway.
Collapse
|
25
|
Hummasti S, Hong C, Bensinger SJ, Tontonoz P. HRASLS3 is a PPARgamma-selective target gene that promotes adipocyte differentiation. J Lipid Res 2008; 49:2535-44. [PMID: 18664718 DOI: 10.1194/jlr.m800269-jlr200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The prevalence of obesity and its associated metabolic diseases worldwide has focused attention on understanding the mechanisms underlying adipogenesis. The nuclear receptor PPARgamma has emerged as a central regulator of adipose tissue function and formation. Despite the identification of numerous PPARgamma targets involved in a range of processes, from lipid droplet formation to adipokine secretion, information is still lacking on targets downstream of PPARgamma that directly affect fat cell differentiation. Here we identify HRASLS3 as a novel PPARgamma regulated gene with a role in adipogenesis. HRASLS3 expression increases during the differentiation of preadipocyte cell lines and is highly expressed in white and brown adipose tissue in mice. HRASLS3 expression is induced by PPARgamma ligands in preadipocyte cell lines as well in adipose tissue in vivo. We demonstrate that the HRASLS3 promoter contains a functional PPAR response element and is a direct target for regulation by PPARgamma/RXR heterodimers. Finally, we show that overexpression of HRASLS3 augments PPARgamma-driven lipid accumulation and adipogenesis, whereas siRNA-mediated knockdown of HRASLS3 expression decreases differentiation. Together, these results identify HRASLS3 as one of the downstream effectors of PPARgamma action in adipogenesis.
Collapse
Affiliation(s)
- Sarah Hummasti
- Howard Hughes Medical Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
26
|
Sáez ME, Grilo A, Morón FJ, Manzano L, Martínez-Larrad MT, González-Pérez A, Serrano-Hernando J, Ruiz A, Ramírez-Lorca R, Serrano-Ríos M. Interaction between Calpain 5, Peroxisome proliferator-activated receptor-gamma and Peroxisome proliferator-activated receptor-delta genes: a polygenic approach to obesity. Cardiovasc Diabetol 2008; 7:23. [PMID: 18657264 PMCID: PMC2527300 DOI: 10.1186/1475-2840-7-23] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Accepted: 07/25/2008] [Indexed: 11/10/2022] Open
Abstract
Context Obesity is a multifactorial disorder, that is, a disease determined by the combined effect of genes and environment. In this context, polygenic approaches are needed. Objective To investigate the possibility of the existence of a crosstalk between the CALPAIN 10 homologue CALPAIN 5 and nuclear receptors of the peroxisome proliferator-activated receptors family. Design Cross-sectional, genetic association study and gene-gene interaction analysis. Subjects The study sample comprise 1953 individuals, 725 obese (defined as body mass index ≥ 30) and 1228 non obese subjects. Results In the monogenic analysis, only the peroxisome proliferator-activated receptor delta (PPARD) gene was associated with obesity (OR = 1.43 [1.04–1.97], p = 0.027). In addition, we have found a significant interaction between CAPN5 and PPARD genes (p = 0.038) that reduces the risk for obesity in a 55%. Conclusion Our results suggest that CAPN5 and PPARD gene products may also interact in vivo.
Collapse
Affiliation(s)
- María E Sáez
- Departamento de Genómica Estructural, Neocodex, C/.,Charles Darwin 6, Acc. A, 41092, Sevilla, Spain.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Maruyama A, Tsukamoto S, Nishikawa K, Yoshida A, Harada N, Motojima K, Ishii T, Nakane A, Yamamoto M, Itoh K. Nrf2 regulates the alternative first exons of CD36 in macrophages through specific antioxidant response elements. Arch Biochem Biophys 2008; 477:139-45. [PMID: 18585365 DOI: 10.1016/j.abb.2008.06.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Revised: 06/07/2008] [Accepted: 06/10/2008] [Indexed: 01/07/2023]
Abstract
We previously demonstrated that Nrf2 regulates oxidized LDL-mediated CD36 expression in macrophages. The current study aimed to determine the mechanism of Nrf2-mediated macrophage CD36 induction. Treatment with the Nrf2 activator diethylmaleate, but not PPARgamma specific ligands, caused marked upregulation of CD36 in mouse macrophage RAW264.7 cells. Similarly, Nrf2 activators induced CD36 expression in bone marrow-derived macrophages in a Nrf2-dependent manner. Induced expression of the three alternative first exons of mouse CD36, deemed 1A, 1B, and 1C, occurred upon Nrf2 activation with exon1A mainly contributing to the CD36 expression. Four antioxidant response elements (AREs) lie within close proximity to these three exons, and chromatin immunoprecipitation assays demonstrated that two AREs upstream of exon1A, the distal 1A-ARE1, and the proximal 1A-ARE2, were Nrf2-responsive. Luciferase reporter assays conclusively demonstrated that 1A-ARE2 is the critical regulatory element for the Nrf2-mediated gene expression. Thus Nrf2 directly regulates CD36 gene expression by binding to 1A-ARE2.
Collapse
Affiliation(s)
- Atsushi Maruyama
- Department of Stress Response Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki 036-8562, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Campbell MJ, Carlberg C, Koeffler HP. A Role for the PPARgamma in Cancer Therapy. PPAR Res 2008; 2008:314974. [PMID: 18528521 PMCID: PMC2408680 DOI: 10.1155/2008/314974] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2008] [Accepted: 03/17/2008] [Indexed: 12/19/2022] Open
Abstract
In 1997, the first published reports highlighted PPARgamma as a novel cancer therapeutic target regulating differentiation of cancer cells. A subsequent flurry of papers described these activities more widely and fuelled further enthusiasm for differentiation therapy, as the ligands for the PPARgamma were seen as well tolerated and in several cases well-established in other therapeutic contexts. This initial enthusiasm and promise was somewhat tempered by contradictory findings in several murine cancer models and equivocal trial findings. As more understanding has emerged in recent years, a renaissance has occurred in targeting PPARgamma within the context of either chemoprevention or chemotherapy. This clarity has arisen in part through a clearer understanding of PPARgamma biology, how the receptor interacts with other proteins and signaling events, and the mechanisms that modulate its transcriptional actions. Equally greater translational understanding of this target has arisen from a clearer understanding of in vivo murine cancer models. Clinical exploitation will most likely require precise and quantifiable description of PPARgamma actions, and resolution of which targets are the most beneficial to target combined with an understanding of the mechanisms that limits its anticancer effectiveness.
Collapse
Affiliation(s)
- Moray J. Campbell
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Carsten Carlberg
- Department of Biosciences, University of Kuopio, 70211 Kuopio, Finland
- Life Sciences Research Unit, University of Luxembourg, 1511 Luxembourg, Luxembourg
| | - H. Phillip Koeffler
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, Los Angeles School of Medicine, University of California, 8700 Beverly Boulevard, Los Angeles, CA 90048, USA
| |
Collapse
|
29
|
Szatmari I, Töröcsik D, Agostini M, Nagy T, Gurnell M, Barta E, Chatterjee K, Nagy L. PPARgamma regulates the function of human dendritic cells primarily by altering lipid metabolism. Blood 2007; 110:3271-80. [PMID: 17664351 DOI: 10.1182/blood-2007-06-096222] [Citation(s) in RCA: 116] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activation of the lipid-regulated nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) modifies the immunophenotype of monocyte-derived dendritic cells (DCs). However it has not been analyzed in a systematic manner how lipid metabolism and immune regulation are connected at the transcriptional level via this receptor. Here we present the genome-wide expression analyses of PPARgamma-instructed human DCs. Receptor activation was achieved by exogenous, synthetic as well as endogenous, natural means. More than 1000 transcripts are regulated during DC development by activation of PPARgamma; half of the changes are positive effects. These changes appear to enhance and modulate the robust gene expression alterations associated with monocyte to DC transition. Strikingly, only genes related to lipid metabolism are overrepresented among early induced genes. As a net consequence, lipid accumulation appears to be diminished in these cells. In contrast, genes related to immune response are regulated after 24 hours, implying the existence of indirect mechanisms of modulation. Receptor dependence was established by using DCs of patients harboring a dominant-negative mutation of PPARgamma. Our data show that PPARgamma acts as a mostly positive transcriptional regulator in human developing DCs, acting primarily through controlling genes involved in lipid metabolism and via this, indirectly modifying the immune phenotype.
Collapse
Affiliation(s)
- Istvan Szatmari
- Department of Biochemistry and Molecular Biology, Research Center for Molecular Medicine, University of Debrecen, Medical and Health Science Center, Nagyerdei krt. 98, Debrecen, Hungary
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Zingarelli B, Hake PW, Mangeshkar P, O'Connor M, Burroughs TJ, Piraino G, Denenberg A, Wong HR. DIVERSE CARDIOPROTECTIVE SIGNALING MECHANISMS OF PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR-γ LIGANDS, 15-DEOXY-Δ12,14-PROSTAGLANDIN J2 AND CIGLITAZONE, IN REPERFUSION INJURY. Shock 2007; 28:554-63. [PMID: 17589386 DOI: 10.1097/shk.0b013e31804f56b9] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a nuclear receptor that regulates diverse biological functions including inflammation. The PPARgamma ligands have been reported to exert cardioprotective effects and attenuate myocardial reperfusion injury. Here, we examined the molecular mechanisms of their anti-inflammatory effects. Male Wistar rats were subjected to myocardial ischemia and reperfusion and were treated with the PPAR-gamma ligands, 15-deoxy-Delta-prostaglandin J2 (15d-PGJ2) or ciglitazone, or with vehicle only, in the absence or presence of the selective PPAR-gamma antagonist GW-9662. In vehicle-treated rats, myocardial injury was associated with elevated tissue activity of myeloperoxidase, indicating infiltration of neutrophils, and elevated plasma levels of creatine kinase and tumor necrosis factor-alpha. These events were preceded by activation of the nuclear factor-kappaB pathway. The PPAR-gamma DNA binding was also increased in the heart after reperfusion. Treatment with ciglitazone or 15d-PGJ2 reduced myocardial damage and neutrophil infiltration and blunted creatine kinase levels and cytokine production. The beneficial effects of both ligands were associated with enhancement of PPAR-gamma DNA binding and reduction of nuclear factor-kappaB activation. Treatment with 15d-PGJ2, but not ciglitazone, enhanced DNA binding of heat shock factor 1 and upregulated the expression of the cardioprotective heat shock protein 70. Treatment with 15d-PGJ2, but not ciglitazone, also induced a significant increase in nuclear phosphorylation of the prosurvival kinase Akt. The cardioprotection afforded by ciglitazone was attenuated by the PPAR-gamma antagonist GW-9662. In contrast, GW-9662 did not affect the beneficial effects afforded by 15d-PGJ2. Thus, our data suggest that treatment with these chemically unrelated PPAR-gamma ligands results in diverse anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Basilia Zingarelli
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, The University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Interleukin-4 mediates the neuroprotective effects of rosiglitazone in the aged brain. Neurobiol Aging 2007; 30:920-31. [PMID: 17950491 DOI: 10.1016/j.neurobiolaging.2007.09.001] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 09/04/2007] [Accepted: 09/05/2007] [Indexed: 12/23/2022]
Abstract
Increased expression of proinflammatory cytokines, like interleukin-1 beta (IL-1 beta), is a feature of the aged brain and it is generally accepted that the primary cell source of these cytokines is activated microglia. In hippocampus of aged rats, the increase in IL-1 beta is accompanied by microglial activation and impaired long-term potentiation (LTP). Peroxisome proliferator-activated receptors (PPARs) possess anti-inflammatory properties that target microglia. In this study the PPAR gamma agonist, rosiglitazone, was orally administered to young and aged rats, and we report that the age-related increases in NO and IL-1 beta production were attenuated in hippocampus of rosiglitazone-treated aged rats and that this was associated with a restoration of LTP. In addition, treatment with rosiglitazone increased interleukin-4 (IL-4) mRNA and reversed the age-related decrease in hippocampal IL-4 concentration. Significantly, while rosiglitazone attenuated the LPS-induced increase in MHCII and IL-1 beta concentration in glia prepared from wildtype mice, it failed to exert an effect in glia prepared from IL-4(-/-) mice, thereby suggesting that the anti-inflammatory actions of rosiglitazone are mediated by its ability to increase IL-4 expression.
Collapse
|
32
|
Jedidi I, Couturier M, Thérond P, Gardès-Albert M, Legrand A, Barouki R, Bonnefont-Rousselot D, Aggerbeck M. Cholesteryl ester hydroperoxides increase macrophage CD36 gene expression via PPARalpha. Biochem Biophys Res Commun 2006; 351:733-8. [PMID: 17084382 DOI: 10.1016/j.bbrc.2006.10.122] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 10/23/2006] [Indexed: 10/23/2022]
Abstract
The uptake of oxidized LDL by macrophages is a key event in the development of atherosclerosis. The scavenger receptor CD36 is one major receptor that internalizes oxidized LDL. In differentiated human macrophages, we compared the regulation of CD36 expression by copper-oxidized LDL or their products. Only oxidized derivatives of cholesteryl ester (CEOOH) increased the amount of CD36 mRNA (2.5-fold). Both oxidized LDL and CEOOH treatment increased two to fourfold the transcription of promoters containing peroxisome-proliferator-activated-receptor responsive elements (PPRE) in the presence of PPARalpha or gamma. Electrophoretic-mobility-shift-assays with nuclear extracts prepared from macrophages treated by either oxidized LDL or CEOOH showed increased binding of PPARalpha to the CD36 gene promoter PPRE. In conclusion, CEOOH present in oxidized LDL increase CD36 gene expression in a pathway involving PPARalpha.
Collapse
Affiliation(s)
- Iness Jedidi
- CNRS, UMR 8601, Laboratoire de Chimie-Physique, Paris F-75006, France
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Larigauderie G, Cuaz-Pérolin C, Younes AB, Furman C, Lasselin C, Copin C, Jaye M, Fruchart JC, Rouis M. Adipophilin increases triglyceride storage in human macrophages by stimulation of biosynthesis and inhibition of beta-oxidation. FEBS J 2006; 273:3498-510. [PMID: 16884492 DOI: 10.1111/j.1742-4658.2006.05357.x] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Lipid accumulation alters macrophage biology and contributes to lipid retention within the vessel wall. In this study, we investigated the role of adipophilin on triglyceride accumulation and lipid-droplet formation in THP-1-derived macrophages (THP-1 macrophages). In the presence of acetylated low-density lipoprotein, macrophages infected with an adenovirus expressing human adipophilin showed a 31% increase in triglyceride content and a greater number of lipid droplets compared with control cells. Incubation of macrophages with very low-density lipoprotein (VLDL) dramatically increased cellular triglyceride content similarly in control and adipophilin-overexpressing cells. By itself, VLDL increased adipophilin expression, which explains the lack of effect of adipophilin overexpression on cellular triglyceride content in macrophages loaded with VLDL. The lipid-droplet content of macrophages was increased by overexpression of adipophilin and/or loading with VLDL. In contrast, inhibition of adipophilin expression using siRNA prevented lipid-droplet formation and significantly reduced intracellular triglyceride content. Using inhibitors of beta-oxidation and acyl-coenzyme A synthetase, results were obtained which suggest that adipophilin elevates cellular lipids by inhibition of beta-oxidation and stimulation of long-chain fatty acid incorporation into triglycerides. Adipophilin expression in THP-1 macrophages altered the cellular content of different lipids and enhanced the size of lipid droplets, consistent with a role for adipophilin in human foam cell formation.
Collapse
|
34
|
Yamaguchi T, Matsushita S, Motojima K, Hirose F, Osumi T. MLDP, a novel PAT family protein localized to lipid droplets and enriched in the heart, is regulated by peroxisome proliferator-activated receptor alpha. J Biol Chem 2006; 281:14232-40. [PMID: 16571721 DOI: 10.1074/jbc.m601682200] [Citation(s) in RCA: 160] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cytosolic lipid droplets (LDs) are multifunctional organelles that exist in all types of eukaryotic cells and control lipid homeostasis. In mammalian cells LDs contain a class of proteins in their surface layers that share a homologous sequence called the PAT domain, including perilipin, adipose differentiation-related protein (ADRP), a tail-interacting protein of 47 kDa (TIP47), and S3-12, which are distributed tissue- or cell type-selectively. Expression in some cases is regulated by peroxisome proliferator-activated receptors (PPARs). In this study we identified a new PAT family member named MLDP (myocardial LD protein) in a murine cDNA data base and showed the mRNA and protein to be highly enriched in the heart and also expressed at lower levels in the liver and adrenals. Upon subcellular fractionation, a substantial amount of MLDP was detected in the top fraction enriched with LDs. Furthermore, overexpressed MLDP tagged with green fluorescent protein accumulated at the surfaces of LDs and co-localized with perilipin and ADRP. Deletion analysis demonstrated the N-terminal region containing a PAT-1 domain and the following 33-mer domain to be required for targeting of MLDP to LDs. MLDP was found to be up-regulated at both mRNA and protein levels in the heart and liver by a selective ligand for PPARalpha, Wy14,643, but not in PPARalpha knock-out mice. MLDP expression was also increased upon fasting in parallel with ADRP. These results indicate that MLDP is a bona fide new PAT family member localized in LDs. Its expression depends on the physiological conditions and the action of PPARalpha.
Collapse
Affiliation(s)
- Tomohiro Yamaguchi
- Graduate School of Life Science, University of Hyogo, 3-2-1 Koto, Kamigori, Hyogo, 678-1297, Japan
| | | | | | | | | |
Collapse
|
35
|
Hughes TA, Stentz F, Gettys T, Smith SR. Combining beta-adrenergic and peroxisome proliferator-activated receptor gamma stimulation improves lipoprotein composition in healthy moderately obese subjects. Metabolism 2006; 55:26-34. [PMID: 16324916 PMCID: PMC2597222 DOI: 10.1016/j.metabol.2005.06.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Accepted: 06/28/2005] [Indexed: 11/21/2022]
Abstract
Current pharmacological regimens for hypertriglyceridemia and low high-density lipoprotein (HDL) are limited to the peroxisome proliferator-activated receptor (PPAR) alpha activating fibrates, niacin, and statins. This pilot study examined the impact of simultaneous stimulation of cyclic adenosine monophosphate with a beta-adrenergic agonist and PPARgamma with pioglitazone (PIO) on lipoprotein composition in moderately obese, healthy subjects. Subjects were treated with PIO (45 mg) to stimulate PPARgamma or a combination of ephedrine (25 mg TID), a beta-agonist, with caffeine (200 mg TID), a phosphodiesterase inhibitor (ephedrine plus caffeine), or both for 16 weeks. Lipoproteins were separated by gradient ultracentrifugation into very low-density lipoprotein (VLDL), intermediate-density lipoprotein, low-density lipoprotein (LDL), and 3 HDL (L, M, and D) subfractions. Apolipoproteins were measured by high-performance liquid chromatography. PIO alone reduced the core triglyceride (TG) content relative to cholesterol ester (CE) in VLDL (-40%), IDL (-25%), and HDL-M (-38%). Ephedrine plus caffeine alone reduced LDL CE (-13%), phospholipids (-9%), and apolipoprotein (apo) B (-13%); increased HDL-M LpA-I (HDL containing apoA-I without apoA-II, 28%), CE/TG (23%), and CE/apoA-I (8%) while reducing apoA-II (-10%); and increased HDL-L LpA-I (29%). Combination therapy reduced total plasma TG (-28%), LDL cholesterol (LDL-C, -10%), apoB (-16%), apoB/apoA-I ratio (-21%) while increasing HDL cholesterol (HDL-C, 21%), total plasma apoA-I (12%), LpA-I (43%), and apoC-I (26%). It also reduced VLDL total mass (-34%) and apoC-III (-39%), LDL CE (-13%), apoB (-13%), and total mass (-11%). Combination therapy increased HDL-L CE/TG (32%), apoC-I (30%), apoA-I (56%), and LpA-I (70%), as well as HDL-M CE (35%), phospholipids (24%), total mass (19%), apoC-I (25%), apoA-I (18%), and LpA-I (56%). In conclusion, simultaneous beta-adrenergic and PPARgamma activation produced beneficial effects on VLDL, LDL, HDL-L, and HDL-M. Perhaps the most important impact of combination therapy was dramatic increases in LpA-I and apoC-I in HDL-L and HDL-M, which were much greater than the sum of the monotherapies. Because LpA-I appears to be the most efficient mediator of reverse-cholesterol transport and a major negative risk factor for cardiovascular disease, this combination therapy may provide very effective treatment of atherosclerosis.
Collapse
Affiliation(s)
- Thomas A Hughes
- Health Science Center, Department of Medicine, University of Tennessee, Memphis, TN 38163, USA.
| | | | | | | |
Collapse
|
36
|
Edvardsson U, Ljungberg A, Lindén D, William-Olsson L, Peilot-Sjögren H, Ahnmark A, Oscarsson J. PPARalpha activation increases triglyceride mass and adipose differentiation-related protein in hepatocytes. J Lipid Res 2005; 47:329-40. [PMID: 16282640 DOI: 10.1194/jlr.m500203-jlr200] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Adipose differentiation-related protein (ADRP) is a lipid droplet-associated protein that is expressed in various tissues. In mice treated with the peroxisome proliferator-activated receptor alpha (PPARalpha) agonist Wy14,643 (Wy), hepatic mRNA and protein levels of ADRP as well as hepatic triglyceride content increased. Also in primary mouse hepatocytes, Wy increased ADRP expression and intracellular triglyceride mass. The triglyceride mass increased in spite of unchanged triglyceride biosynthesis and increased palmitic acid oxidation. However, Wy incubation decreased the secretion of newly synthesized triglycerides, whereas apolipoprotein B secretion increased. Thus, decreased availability of triglycerides for VLDL assembly could help to explain the cellular accumulation of triglycerides after Wy treatment. We hypothesized that this effect could be mediated by increased ADRP expression. Similar to PPARalpha activation, adenovirus-mediated ADRP overexpression in mouse hepatocytes enhanced cellular triglyceride mass and decreased the secretion of newly synthesized triglycerides. In ADRP-overexpressing cells, Wy incubation resulted in a further decrease in triglyceride secretion. This effect of Wy was not attributable to decreased cellular triglycerides after increased fatty acid oxidation because the triglyceride mass in Wy-treated ADRP-overexpressing cells was unchanged. In summary, PPARalpha activation prevents the availability of triglycerides for VLDL assembly and increases hepatic triglyceride content in part by increasing the expression of ADRP.
Collapse
Affiliation(s)
- Ulrika Edvardsson
- Wallenberg Laboratory for Cardiovascular Research, Sahlgrenska University Hospital, Göteborg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
37
|
Kelly K, Hutchinson G, Nebenius-Oosthuizen D, Smith AJH, Bartsch JW, Horiuchi K, Rittger A, Manova K, Docherty AJP, Blobel CP. Metalloprotease-disintegrin ADAM8: expression analysis and targeted deletion in mice. Dev Dyn 2005; 232:221-31. [PMID: 15580619 DOI: 10.1002/dvdy.20221] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
ADAM8 (a disintegrin and metalloprotease 8, also referred to as MS2/CD156a) is a membrane-anchored metalloprotease that was first identified in a macrophage cell line and has been implicated in neurodegenerative diseases. Here, we evaluated the expression of ADAM8 during mouse development and generated mice lacking ADAM8 (Adam8-/- mice). During early mouse development, ADAM8 is expressed by maternal cells in the decidua and by trophoblast derivatives of the embryo but not in the derivatives of the inner cell mass. At later stages, prominent expression of ADAM8 is seen in the embryo proper, in the gonadal ridge, thymus, developing cartilage and bone, brain and spinal cord, and in the mesenchyme in close proximity to the branch point between the jugular vein and developing lymphatic vessels. Examination of Adam8-/- mice, however, revealed no major defects in these or other structures during development or in adult tissues and no evident pathological phenotypes.
Collapse
Affiliation(s)
- Kristine Kelly
- Cell Biology Program, Sloan-Kettering Institute, New York, New York, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
McGuire K, Glass EJ. The expanding role of microarrays in the investigation of macrophage responses to pathogens. Vet Immunol Immunopathol 2005; 105:259-75. [PMID: 15808305 DOI: 10.1016/j.vetimm.2005.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
In the last few years, microarray technology has emerged as the method of choice for large-scale gene expression studies. It provides an efficient and rapid method to investigate the entire transcriptome of a cell. No research field has benefited more from microarray technology than the study of the exquisite interplay between pathogens and hosts. Numerous microarray studies have now been published in this field, which have provided insights into the mechanisms of host defence and the tactics employed by pathogens to circumvent these protection strategies. These studies have led to a more comprehensive understanding of the host immune response and identified new avenues of research for potential control strategies against pathogens. In the past, research has concentrated on human and mouse microarrays to investigate host-pathogen interactions, regardless of the host species. This trend is changing with the ever-expanding sequence resources now available for many pathogen and host species, including livestock animals. The use of species-specific microarrays has furthered our understanding of host-pathogen interactions for particular organisms and aided in the annotation of unknown genes. Macrophages play a central role in the host's innate and adaptive immune responses to pathogens. These cells are in the first line of defence and interact with a wide range of pathogens; many of which have evolved strategies to circumvent the macrophage defence mechanisms and survive within these cells. In this report, we review the wealth of studies using microarray technology to investigate the response of macrophages to pathogens. These studies illustrate how microarray technology has expanded our understanding of the dialogue between macrophage and pathogen and provide examples of the benefits and pitfalls of using this technique. Furthermore, we discuss the resources available to use microarray analysis to study the immune response of a non-human, non-rodent species, the cow.
Collapse
Affiliation(s)
- Kirsty McGuire
- Department of Genetics and Genomics, Roslin Institute, Roslin, Midlothian, Edinburgh EH25 9PS, UK.
| | | |
Collapse
|
39
|
Chien CL, Chen YC, Chang MF, Greenberg AS, Wang SM. Magnolol induces the distributional changes of p160 and adipose differentiation-related protein in adrenal cells. Histochem Cell Biol 2005; 123:429-39. [PMID: 15844002 DOI: 10.1007/s00418-005-0771-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2005] [Indexed: 11/26/2022]
Abstract
Magnolol stimulates adrenal steroidogenesis and induces the distributional changes of p160 and adipose differentiation-related protein (ADRP) in rat adrenal cells. This study investigated the underlying signaling mechanisms involved in these processes. Magnolol (30 microM) caused a time-dependent increase in the phosphorylation of extracellular signal-related kinase (ERK) in cultured adrenal cells. The following evidence supports a link between ERK activation and p160 translocation. First, the magnolol-induced redistribution of p160 from the lipid droplet surface to the cytosol, resulting in the decrease in the percentages of p160-positive cells, and this decrease in p160-positive cells was completely blocked by pretreatment with either of the MAPK-ERK kinase (MEK) inhibitors PD98059 or U0126. Second, magnolol did not significantly decrease total p160 protein levels but caused an increase in threonine phosphorylation of p160, which reached a maximum after 5 min of magnolol treatment, and this magnolol-induced phosphorylation of p160 was prevented by pretreatment with U0126, suggesting the involvement of ERK. In addition, magnolol decreased both ADRP immunostaining intensity at the lipid droplet surface and the percentage of ADRP-positive cells. This was further confirmed biochemically by the decrease in ADRP levels in total cell homogenates and in lipid droplet fractions. Magnolol-induced decrease in ADRP staining at the lipid droplet surface was not affected by pretreatment with PD98059 or U0126, indicating that ERK signaling was not involved in this event. Furthermore, treatment with 30 microM magnolol for 6 h resulted in about 50% decrease in ADRP protein level. Therefore, decreased protein levels of p160 and ADRP at the lipid droplet surface induced by magnolol were mediated via two different mechanisms: phosphorylation of p160 and downregulation of ADRP expression, respectively.
Collapse
Affiliation(s)
- Chung-Liang Chien
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, 1-1 Jen-Ai Road, Taipei, 100, Taiwan
| | | | | | | | | |
Collapse
|
40
|
Shiraki T, Kamiya N, Shiki S, Kodama TS, Kakizuka A, Jingami H. α,β-Unsaturated Ketone Is a Core Moiety of Natural Ligands for Covalent Binding to Peroxisome Proliferator-activated Receptor γ. J Biol Chem 2005; 280:14145-53. [PMID: 15695504 DOI: 10.1074/jbc.m500901200] [Citation(s) in RCA: 186] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma) functions in various biological processes, including macrophage and adipocyte differentiation. Several natural lipid metabolites have been shown to activate PPARgamma. Here, we report that some PPARgamma ligands, including 15-deoxy-Delta12,14-prostaglandin J2, covalently bind to a cysteine residue in the PPARgamma ligand binding pocket through a Michael addition reaction by an alpha,beta-unsaturated ketone. Using rhodamine-maleimide as well as mass spectroscopy, we showed that the binding of these ligands is covalent and irreversible. Consistently, mutation at the cysteine residue abolished abilities of these ligands to activate PPARgamma, but not of BRL49653, a non-covalent synthetic agonist, indicating that covalent binding of the alpha,beta-unsaturated ketone in the natural ligands was required for their transcriptional activities. Screening of lipid metabolites containing the alpha,beta-unsaturated ketone revealed that several other oxidized metabolites of hydroxyeicosatetraenoic acid, hydroxyeicosadecaenoic acid, and prostaglandins can also function as novel covalent ligands for PPARgamma. We propose that PPARgamma senses oxidation of fatty acids by recognizing such an alpha,beta-unsaturated ketone as a common moiety.
Collapse
Affiliation(s)
- Takuma Shiraki
- Department of Molecular Biology, Biomolecular Engineering Research Institute, 6-2-3 Furuedai, Suita-City, Osaka 565-0874, Japan
| | | | | | | | | | | |
Collapse
|
41
|
Matthiessen MW, Pedersen G, Albrektsen T, Adamsen S, Fleckner J, Brynskov J. Peroxisome proliferator-activated receptor expression and activation in normal human colonic epithelial cells and tubular adenomas. Scand J Gastroenterol 2005; 40:198-205. [PMID: 15764152 DOI: 10.1080/00365520410009573] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Peroxisome proliferator-activated receptor (PPAR) ligands, widely used in type 2 diabetes treatment, have variably been shown to promote or prevent colon tumor formation in animal models and cell lines, but their role in normal human colon is unknown. The aim of this study was to determine PPAR expression and function in normal human colonic epithelial cells and tubular adenomas. MATERIAL AND METHODS Short-term cultures of normal human colonic epithelial cells were established from biopsies obtained in 42 patients with normal colonoscopy. PPAR and adipophilin mRNA expression was assessed by real-time RT-PCR. PPARs were activated by ligands for PPAR alpha (Wy-14643), PPAR delta (GW-501516) and PPAR gamma (rosiglitazone or troglitazone). Cell viability was measured using the methyltetrazoleum assay, proliferation by thymidine incorporation, and DNA profiles by flow cytometry. PPAR mRNA levels in tubular adenomas or metaplastic polyps (n=12) were compared with those in controls. RESULTS PPAR alpha and gamma were consistently expressed in normal colonocytes while no PPAR delta expression could be detected. PPAR gamma activation induced a 7.5-fold increase in adipophilin expression (a PPAR-activated gene). PPAR gamma activation had no effect on viability or DNA profiles, but led to a 25% significant decrease in cell proliferation. Finally, a selective and significant 2.5-fold decrease in PPAR alpha expression was observed in tubular adenomas, but not in metaplastic polyps, compared to controls. CONCLUSIONS Our findings support the view that PPAR gamma ligands act as anti-proliferative agents rather than as promoters of tumorigenesis in normal human colon. Moreover, they raise interest in investigation of PPAR alpha as a therapeutic target to prevent adenoma formation.
Collapse
|
42
|
Llaverias G, Lacasa D, Viñals M, Vázquez-Carrera M, Sánchez RM, Laguna JC, Alegret M. Reduction of intracellular cholesterol accumulation in THP-1 macrophages by a combination of rosiglitazone and atorvastatin. Biochem Pharmacol 2004; 68:155-63. [PMID: 15183127 DOI: 10.1016/j.bcp.2004.03.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2003] [Accepted: 03/19/2004] [Indexed: 11/21/2022]
Abstract
Rosiglitazone and atorvastatin combination therapy has beneficial effects on both glycemic control and plasma lipid levels in type 2 diabetic patients. In the present study, we sought to determine whether this combination can also exert direct antiatherosclerotic effects in macrophages. Our results show that 2 microM rosiglitazone, alone or combined with 5 microM atorvastatin, significantly upregulated the expression of the ATP-binding cassette transporter ABCA1 and of the class B scavenger receptor CLA-1 (CD36 and LIMPII analog), both involved in cholesterol efflux from macrophages. On the other hand, the combination with atorvastatin attenuated the inductive response elicited by rosiglitazone alone on CD36 mRNA (34%, P < 0.05) and protein (16%, P < 0.05), while the uptake of oxidized low density lipoprotein (LDL) remained unaffected. When we examined the effects of the drugs on acetyl-LDL-induced cholesterol accumulation, we found that only the combination of atorvastatin with rosiglitazone caused a net depletion in the cholesteryl ester content of macrophages (35%, P < 0.05). Our data suggest that this reduction was not mediated by effects on proteins that regulate cholesterol flux, but it may be related to the inhibition of cholesteryl ester formation elicited by the statin.
Collapse
Affiliation(s)
- Gemma Llaverias
- Unitat de Farmacologia, Departament de Farmacologia i Química Terapèutica, Facultat de Farmàcia, Universitat de Barcelona, Barcelona 08028, Spain
| | | | | | | | | | | | | |
Collapse
|