1
|
Pan R, Liu J, Chen Y. Treatment of obesity-related diabetes: significance of thermogenic adipose tissue and targetable receptors. Front Pharmacol 2023; 14:1144918. [PMID: 37435495 PMCID: PMC10332465 DOI: 10.3389/fphar.2023.1144918] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/12/2023] [Indexed: 07/13/2023] Open
Abstract
Diabetes mellitus is mainly classified into four types according to its pathogenesis, of which type 2 diabetes mellitus (T2DM) has the highest incidence rate and is most relevant to obesity. It is characterized by high blood glucose, which is primarily due to insulin resistance in tissues that are responsible for glucose homeostasis (such as the liver, skeletal muscle, and white adipose tissue (WAT)) combined with insufficiency of insulin secretion from pancreatic β-cells. Treatment of diabetes, especially treatment of diabetic complications (such as diabetic nephropathy), remains problematic. Obesity is one of the main causes of insulin resistance, which, however, could potentially be treated by activating thermogenic adipose tissues, like brown and beige adipose tissues, because they convert energy into heat through non-shivering thermogenesis and contribute to metabolic homeostasis. In this review, we summarize the function of certain anti-diabetic medications with known thermogenic mechanisms and focus on various receptor signaling pathways, such as previously well-known and recently discovered ones that are involved in adipose tissue-mediated thermogenesis and could be potentially targeted to combat obesity and its associated diabetes, for a better understanding of the molecular mechanisms of non-shivering thermogenesis and the development of novel therapeutic interventions for obesity-related diabetes and potentially diabetic complications.
Collapse
Affiliation(s)
- Ruping Pan
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiadai Liu
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Endocrinology and Metabolism, Ministry of Education, Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Chen
- Department of Endocrinology, Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Endocrinology and Metabolism, Ministry of Education, Key Laboratory of Vascular Aging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan, Hubei, China
| |
Collapse
|
2
|
Yousefipour Z, Chug N, Marek K, Nesbary A, Mathew J, Ranganna K, Newaz MA. Contribution of PPARγ in modulation of acrolein-induced inflammatory signaling in gp91 phox knock-out mice. Biochem Cell Biol 2017; 95:482-490. [PMID: 28376311 DOI: 10.1139/bcb-2016-0198] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Oxidative stress and inflammation are major contributors to acrolein toxicity. Peroxisome proliferator activated receptor gamma (PPARγ) has antioxidant and anti-inflammatory effects. We investigated the contribution of PPARγ ligand GW1929 to the attenuation of oxidative stress in acrolein-induced insult. Male gp91phox knock-out (KO) mice were treated with acrolein (0.5 mg·(kg body mass)-1 by intraperitoneal injection for 7 days) with or without GW1929 (GW; 0.5 mg·(kg body mass)-1·day-1, orally, for 10 days). The livers were processed for further analyses. Acrolein significantly increased 8-isoprostane and reduced PPARγ activity (P < 0.05) in the wild type (WT) and KO mice. GW1929 reduced 8-isoprostane (by 32% and 40% in WT and KO mice, respectively) and increased PPARγ activity (by 81% and 92% in WT and KO, respectively). Chemokine activity was increased (by 63%) in acrolein-treated WT mice, and was reduced by GW1929 (by 65%). KO mice exhibited higher xanthine oxidase (XO). Acrolein increased XO and COX in WT mice and XO in KO mice. GW1929 significantly reduced COX in WT and KO mice and reduced XO in KO mice. Acrolein significantly reduced the total antioxidant status in WT and KO mice (P < 0.05), which was improved by GW1929 (by 75% and 74%). The levels of NF-κB were higher in acrolein-treated WT mice. GW1929 reduced NF-κB levels (by 51%) in KO mice. Acrolein increased CD36 in KO mice (by 43%), which was blunted with GW1929. Data confirms that the generation of free radicals by acrolein is mainly through NAD(P)H, but other oxygenates play a role too. GW1929 may alleviate the toxicity of acrolein by attenuating NF-κB, COX, and CD36.
Collapse
Affiliation(s)
- Zivar Yousefipour
- a College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Neha Chug
- b College of Pharmacy, Chicago State University, Chicago, IL 60503, USA
| | - Katarzyna Marek
- b College of Pharmacy, Chicago State University, Chicago, IL 60503, USA
| | - Alicia Nesbary
- b College of Pharmacy, Chicago State University, Chicago, IL 60503, USA
| | - Joseph Mathew
- a College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Kasturi Ranganna
- a College of Pharmacy & Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | - Mohammad A Newaz
- b College of Pharmacy, Chicago State University, Chicago, IL 60503, USA
| |
Collapse
|
3
|
Chow YL, Sogame M, Sato F. 13-Methylberberine, a berberine analogue with stronger anti-adipogenic effects on mouse 3T3-L1 cells. Sci Rep 2016; 6:38129. [PMID: 27917887 PMCID: PMC5137028 DOI: 10.1038/srep38129] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 11/04/2016] [Indexed: 12/03/2022] Open
Abstract
Lipid metabolism modulation is a main focus of metabolic syndrome research, an area in which many natural and synthetic chemicals are constantly being screened for in vitro and in vivo activity. Berberine, a benzylisoquinoline plant alkaloid, has been extensively investigated for its anti-obesity effects and as a potential cholesterol and triglyceride-lowering drug. We screened 11 protoberberine and 2 benzophenanthridine alkaloids for their anti-adipogenic effects on 3T3-L1 adipocytes and found that 13-methylberberine exhibited the most potent activity. 13-Methylberberine down-regulated the expression of the main adipocyte differentiation transcription factors, peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT enhancer binding protein alpha (C/EBPα), as well as their target genes. PPARγ, C/EBPα, and sterol regulatory element binding protein 1 (SREBP-1) protein levels were reduced, and this lipid-reducing effect was attenuated by an AMP-activated protein kinase (AMPK) inhibitor, indicating that the effect of this compound requires the AMPK signaling pathway. Decreased Akt phosphorylation suggested reduced de novo lipid synthesis. C-13 methyl substitution of berberine increased its accumulation in treated cells, suggesting that 13-methylberberine has improved absorption and higher accumulation compared to berberine. Our findings suggest that 13-methylberberine has potential as an anti-obesity drug.
Collapse
Affiliation(s)
- Yit-Lai Chow
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kitashirakawa, Sakyo, Kyoto, 606-8502, Japan
| | - Mami Sogame
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kitashirakawa, Sakyo, Kyoto, 606-8502, Japan
| | - Fumihiko Sato
- Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kitashirakawa, Sakyo, Kyoto, 606-8502, Japan
| |
Collapse
|
4
|
Chang CW, Abhinav K, Di Cara F, Panagakou I, Vass S, Heck MMS. A role for the metalloprotease invadolysin in insulin signaling and adipogenesis. Biol Chem 2016; 398:373-393. [PMID: 27622830 DOI: 10.1515/hsz-2016-0226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 09/04/2016] [Indexed: 01/24/2023]
Abstract
Invadolysin is a novel metalloprotease conserved amongst metazoans that is essential for life in Drosophila. We previously showed that invadolysin was essential for the cell cycle and cell migration, linking to metabolism through a role in lipid storage and interaction with mitochondrial proteins. In this study we demonstrate that invadolysin mutants exhibit increased autophagy and decreased glycogen storage - suggestive of a role for invadolysin in insulin signaling in Drosophila. Consistent with this, effectors of insulin signaling were decreased in invadolysin mutants. In addition, we discovered that invadolysin was deposited on newly synthesized lipid droplets in a PKC-dependent manner. We examined two in vitro models of adipogenesis for the expression and localization of invadolysin. The level of invadolysin increased during both murine 3T3-L1 and human Simpson-Golabi-Behmel syndrome (SGBS), adipogenesis. Invadolysin displayed a dynamic localization to lipid droplets over the course of adipogenesis, which may be due to the differential expression of distinct invadolysin variants. Pharmacological inhibition of adipogenesis abrogated the increase in invadolysin. In summary, our results on in vivo and in vitro systems highlight an important role for invadolysin in insulin signaling and adipogenesis.
Collapse
|
5
|
Dose apolipoprotein AV has influence on the regulation of fatty acids and triglyceride metabolism in cardiomyocyte in case of obesity. Int J Cardiol 2015; 185:109-11. [PMID: 25791104 DOI: 10.1016/j.ijcard.2015.03.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 03/07/2015] [Indexed: 11/23/2022]
|
6
|
Krist B, Florczyk U, Pietraszek-Gremplewicz K, Józkowicz A, Dulak J. The Role of miR-378a in Metabolism, Angiogenesis, and Muscle Biology. Int J Endocrinol 2015; 2015:281756. [PMID: 26839547 PMCID: PMC4709675 DOI: 10.1155/2015/281756] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 11/30/2015] [Indexed: 02/06/2023] Open
Abstract
MicroRNA-378a (miR-378a, previously known as miR-378) is one of the small noncoding RNA molecules able to regulate gene expression at posttranscriptional level. Its two mature strands, miR-378a-3p and miR-378a-5p, originate from the first intron of the peroxisome proliferator-activated receptor gamma, coactivator 1 beta (ppargc1b) gene encoding PGC-1β. Embedding in the sequence of this transcriptional regulator of oxidative energy metabolism implies involvement of miR-378a in metabolic pathways, mitochondrial energy homeostasis, and related biological processes such as muscle development, differentiation, and regeneration. On the other hand, modulating the expression of proangiogenic factors such as vascular endothelial growth factor, angiopoietin-1, or interleukin-8, influencing inflammatory reaction, and affecting tumor suppressors, such as SuFu and Fus-1, miR-378a is considered as a part of an angiogenic network in tumors. In the latter, miR-378a can evoke broader actions by enhancing cell survival, reducing apoptosis, and promoting cell migration and invasion. This review describes the current knowledge on miR-378a linking oxidative/lipid metabolism, muscle biology, and blood vessel formation.
Collapse
Affiliation(s)
- Bart Krist
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Urszula Florczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Katarzyna Pietraszek-Gremplewicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Alicja Józkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30–387 Krakow, Poland
- *Jozef Dulak:
| |
Collapse
|
7
|
Hagberg C, Mehlem A, Falkevall A, Muhl L, Eriksson U. Endothelial fatty acid transport: role of vascular endothelial growth factor B. Physiology (Bethesda) 2014; 28:125-34. [PMID: 23455771 DOI: 10.1152/physiol.00042.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Dietary lipids present in the circulation have to be transported through the vascular endothelium to be utilized by tissue cells, a vital mechanism that is still poorly understood. Vascular endothelial growth factor B (VEGF-B) regulates this process by controlling the expression of endothelial fatty acid transporter proteins (FATPs). Here, we summarize research on the role of the vascular endothelium in nutrient transport, with emphasis on VEGF-B signaling.
Collapse
|
8
|
Mansour M. The Roles of Peroxisome Proliferator-Activated Receptors in the Metabolic Syndrome. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 121:217-66. [DOI: 10.1016/b978-0-12-800101-1.00007-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
9
|
The Korean Mistletoe (Viscum album coloratum) Extract Has an Antiobesity Effect and Protects against Hepatic Steatosis in Mice with High-Fat Diet-Induced Obesity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:168207. [PMID: 23935653 PMCID: PMC3725881 DOI: 10.1155/2013/168207] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 04/14/2013] [Indexed: 01/07/2023]
Abstract
This study investigates the inhibitory effects of Korean mistletoe extract (KME) on adipogenic factors in 3T3-L1 cells and obesity and nonalcoholic fatty liver disease (NAFLD) in mice fed a high-fat diet. Male C57Bl/6 mice fed a high-fat diet were treated with KME (3 g/kg/day) for 15 weeks for the antiobesity and NAFLD experiments. Body weight and daily food intake were measured regularly during the experimental period. The epididymal pad was measured and liver histology was observed. The effects of KME on thermogenesis and endurance capacity were measured. The effects of KME on adipogenic factors were examined in 3T3-L1 cells. Body and epididymal fat pad weights were reduced in KME-treated mice, and histological examination showed an amelioration of fatty liver in KME-treated mice, without an effect on food consumption. KME potently induces mitochondrial activity by activating thermogenesis and improving endurance capacity. KME also inhibited adipogenic factors in vitro. These results demonstrate the inhibitory effects of KME on obesity and NAFLD in mice fed a high-fat diet. The effects appear to be mediated through an enhanced mitochondrial activity. Therefore, KME may be an effective therapeutic candidate for treating obesity and fatty liver caused by a high-fat diet.
Collapse
|
10
|
Zhu HJ, Ding HH, Deng JY, Pan H, Wang LJ, Li NS, Wang XQ, Shi YF, Gong FY. Inhibition of preadipocyte differentiation and adipogenesis by zinc-α2-glycoprotein treatment in 3T3-L1 cells. J Diabetes Investig 2013; 4:252-60. [PMID: 24843663 PMCID: PMC4015661 DOI: 10.1111/jdi.12046] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/29/2012] [Accepted: 11/27/2012] [Indexed: 12/19/2022] Open
Abstract
Aims/Introduction Zinc‐α2‐glycoprotein (ZAG) is associated with the loss of adipose tissue in cancer cachexia, and has recently been proposed to be a candidate factor in the regulation of bodyweight. The aim of the study was to investigate the effects of ZAG on the proliferation and differentiation of 3T3‐L1 preadipocytes. Materials and Methods 3‐(4,5‐Dimethylthiazol‐2‐yl) 2,5‐diphenyl tetrazolium bromide (MTT) spectrophotometry, Oil Red O staining, intracellular triglyceride assays, real‐time quantitative reverse transcription polymerase chain reaction and transient transfection methods were used to explore the action of ZAG. Results Ectopic ZAG expression significantly stimulates 3T3‐L1 cells proliferation in a dose‐ and time‐dependent manner. The maximum influence of ZAG on proliferation was 1.43‐fold higher than what was observed in control cells. This effect was observed 144 h after transfection with 0.16 μg of murine ZAG (mZAG) plasmid (P < 0.001). The intracellular lipids content in mZAG over‐expressing cells were decreased as much as 37% when compared with the control cells after differentiation (P < 0.05, P < 0.01). The messenger ribonucleic acid levels of peroxisome proliferators‐activated receptor‐γ (PPARγ), CCAAT enhancer‐binding protein‐α (C/EBPα) and the critical lipogenic gene, fatty acid synthase (FAS), are also downregulated by up to 50% in fully differentiated ZAG‐treated adipocytes. ZAG suppresses FAS messenger ribonucleic acid expression by reducing FAS promoter activity. Conclusions Zinc‐α2‐glycoprotein stimulates the proliferation and inhibits the differentiation of 3T3‐L1 murine preadipocytes. The inhibitory action of ZAG on cell differentiation might be a result of the attenuation of the expression of PPARγ, C/EBPα and the lipogenic‐specific enzyme FAS by reducing FAS promoter activity.
Collapse
Affiliation(s)
- Hui-Juan Zhu
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Hui-Hua Ding
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China ; Department of Rheumatology Renji Hospital, School of Medicine Shanghai Jiao Tong University Shanghai China
| | - Jie-Ying Deng
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Hui Pan
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Lin-Jie Wang
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Nai-Shi Li
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Xiang-Qing Wang
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Yi-Fan Shi
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Feng-Ying Gong
- Department of Endocrinology Key Laboratory of Endocrinology of Ministry of Health Peking Union Medical College Hospital Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| |
Collapse
|
11
|
Yadav S, Anbalagan M, Shi Y, Wang F, Wang H. Arsenic inhibits the adipogenic differentiation of mesenchymal stem cells by down-regulating peroxisome proliferator-activated receptor gamma and CCAAT enhancer-binding proteins. Toxicol In Vitro 2012; 27:211-9. [PMID: 23108036 DOI: 10.1016/j.tiv.2012.10.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 11/24/2022]
Abstract
Arsenic remains a top environmental concern in the United States as well as worldwide because of its global existence and serious health impacts. Apoptotic effect of arsenic in human mesenchymal stem cells (hMSCs) has been identified in our previous study; the effects of arsenic on hMSCs remain largely unknown. Here, we report that arsenic inhibits the adipogenic differentiation of human mesenchymal stem cells (hMSCs). Arsenic reduced the formation of lipid droplets and the expression of adipogenesis-related proteins, such as CCAAT enhancer binding protein-(C/EBPs), peroxisome proliferator-activated receptor-gamma (PPAR-γ), and adipocyte fatty acid-binding protein aP2 (aP2). Arsenic mediates this process by sustaining PPAR-γ activity. In addition, inhibition of PPAR-γ activity with T0070907 and up-regulation with its agonist troglitazone, showed the direct association of PPAR-γ and arsenic-mediated inhibition of differentiating hMSCs. Taken together, these results indicate that arsenic inhibits adipogenic differentiation through PPAR-γ pathway and suggest a novel inhibitory effect of arsenic on adipogenic differentiation in hMSCs.
Collapse
Affiliation(s)
- Santosh Yadav
- Tulane Cancer Center, Tulane University, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
12
|
Adipocyte pyruvate dehydrogenase kinase 4 expression is associated with augmented PPARγ upregulation in early-life programming of later obesity. FEBS Open Bio 2012; 2:32-6. [PMID: 23650578 PMCID: PMC3642103 DOI: 10.1016/j.fob.2012.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 02/14/2012] [Accepted: 02/17/2012] [Indexed: 01/08/2023] Open
Abstract
We studied adipocytes from 8-week-old control rat offspring (CON) or rat offspring subjected to maternal low (8%) protein (MLP) feeding during pregnancy/lactation, a procedure predisposing to obesity. Acute exposure to isoproterenol or adenosine enhanced PDK4 and PPARγ mRNA gene expression in CON and MLP adipocytes. Enhanced adipocyte Pdk4 expression correlated with increased PPARγ expression. Higher levels of PDK4 and PPARγ were observed in MLP adipocytes. SCD1 is a PPARγ target. Isoproterenol enhanced adipocyte PDK4 and SCD1 gene expression in parallel. This could reflect augmented PPARγ expression together with enhanced lipolytic stimulation to supply endogenous PPARγ ligands, allowing enhanced adipocyte PDK4 and SCD1 expression via PPARγ activation. In contrast, the effect of adenosine to increase PDK4 expression is independent of stimulation of lipolysis and, as SCD1 expression was unaffected by adenosine, unlikely to reflect PPARγ activation. Increased adipocyte expression of both PDK4 and SCD1 in the MLP model could participate as components of a "thrifty" phenotype, favouring the development of obesity.
Collapse
Key Words
- ADO, adenosine
- Adipose tissue
- BSA, bovine serum albumin
- CON, control
- HSL, hormone-sensitive lipase
- ISO, isoproterenol
- KRHB, Krebs–Ringer HEPES buffer
- Lipogenesis
- MLP, maternal low protein
- NEFA, non-esterified fatty acid
- PC, pyruvate carboxylase
- PDC, pyruvate dehydrogenase complex
- PDK, pyruvate dehydrogenase kinase
- PEPCK, phosphoenolpyruvate carboxykinase
- PPAR, peroxisome proliferator-activated receptor
- Programming
- Pyruvate dehydrogenase complex
- SCD, stearoyl-CoA desaturase
- TAG, triacylglycerol
- WAT, white adipose tissue
Collapse
|
13
|
Hwang HS, Kim SH, Yoo YG, Chu YS, Shon YH, Nam KS, Yun JW. Inhibitory effect of deep-sea water on differentiation of 3T3-L1 adipocytes. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2009; 11:161-168. [PMID: 18654820 DOI: 10.1007/s10126-008-9131-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2008] [Accepted: 07/04/2008] [Indexed: 05/26/2023]
Abstract
Currently, the utilization of deep-sea water (DSW) is receiving much attention due to its high productivity, large quantity, and potential for biological application. The 3T3-L1 cell line is a well-established and commonly used in vitro model to assess adipocyte differentiation. Over the course of several days, confluent 3T3-L1 cells can be converted to adipocytes in the presence of an adipogenic cocktail. In this study, the effects of DSW on differentiation adipocyte 3T3-L1 cells were studied. DSW significantly decreased lipid accumulation, a marker of adipogenesis, in a dose-dependent manner. DSW of hardness 1,000 was the most effective for inhibiting adipocyte differentiation without any cytotoxicity. DSW significantly reduced expression mRNA levels of PPARgamma and C/EBPalpha and protein levels of fatty-acid-binding protein and adiponectin. Our results suggest a potential role for DSW as anti-obesity agents by inhibiting adipocyte differentiation mediated through the down-regulated expression of adipogenic transcription factors and adipocyte-specific proteins.
Collapse
Affiliation(s)
- Hee Sun Hwang
- Department of Biotechnology, Daegu University, Kyungbuk, Kyungsan, 712-714, South Korea
| | | | | | | | | | | | | |
Collapse
|
14
|
A novel peroxisome proliferator-activated receptor alpha/gamma agonist, BPR1H0101, inhibits topoisomerase II catalytic activity in human cancer cells. Anticancer Drugs 2008; 19:151-8. [PMID: 18176111 DOI: 10.1097/cad.0b013e3282f28fe] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Peroxisome proliferator-activated receptor (PPAR) gamma agonists are used clinically for treating diabetes mellitus and cancer. 2-Methyl-2[(1-{3-phenyl-7-propylbenzol[d]isoxazol-6-yl}oxy)propyl]-1H-4-indolyl) oxy]propanoic acid (BPR1H0101) is a novel synthetic indole-based compound, discovered through research to identify new PPARgamma agonists, and it acts as a dual agonist for PPARgamma and PPARalpha. Isobologram analysis demonstrated that BPR1H0101 is capable of antagonistic interaction with the topoisomerase (topo) II poison, VP16. A study of its mechanism showed that BPR1H0101 could inhibit the catalytic activity of topo II in vitro, but did not produce detectable topo II-mediated DNA strand breaks in human oral cancer KB cells. Furthermore, BPR1H0101 could inhibit VP16-induced topo II-mediated DNA cleavage and ataxia-telangiectasia-mutated phosphorylation in KB cells. The results suggest that BPR1H0101 can interfere with the topo II reaction by inhibiting catalytic activity before the formation of the intermediate cleavable complex; consequently, it can impede VP16-induced topo II-mediated DNA cleavage and cell death. This is the first identified PPARalpha/gamma agonist that can serve as a topo II catalytic inhibitor, to interfere with VP16-induced cell death. The result might have relevance to the clinical use of the PPARalpha/gamma agonist in combination chemotherapy.
Collapse
|
15
|
Abstract
Alzheimer disease (AD) is defined by progressive impairments in memory and cognition and by the presence of extracellular neuritic plaques and intracellular neurofibrillary tangles. However, oxidative stress and impaired mitochondrial function always accompany AD. Mitochondria are a major site of production of free radicals [ie, reactive oxygen species (ROS)] and primary targets of ROS. ROS are cytotoxic, and evidence of ROS-induced damage to cell membranes, proteins, and DNA in AD is overwhelming. Nevertheless, therapies based on antioxidants have been disappointing. Thus, alternative strategies are necessary. ROS also act as signaling molecules including for transcription. Thus, chronic exposure to ROS in AD could activate cascades of genes. Although initially protective, prolonged activation may be damaging. Thus, therapeutic approaches based on modulation of these gene cascades may lead to effective therapies. Genes involved in several pathways including antioxidant defense, detoxification, inflammation, etc, are induced in response to oxidative stress and in AD. However, genes that are associated with energy metabolism, which is necessary for normal brain function, are mostly down-regulated. Redox-sensitive transcription factors such as activator protein-1, nuclear factor-kappaB, specificity protein-1, and hypoxia-inducible factor are important in redox-dependent gene regulation. Peroxisome proliferators-activated receptor-gamma coactivator (PGC-1alpha) is a coactivator of several transcription factors and is a potent stimulator of mitochondrial biogenesis and respiration. Down-regulated expression of PGC-1alpha has been implicated in Huntington disease and in several Huntington disease animal models. PGC-1alpha role in regulation of ROS metabolism makes it a potential candidate player between ROS, mitochondria, and neurodegenerative diseases. This review summarizes the current progress on how oxidative stress regulates the expression of genes that might contribute to AD pathophysiology and the implications of the transcriptional modifications for AD. Finally, potential therapeutic strategies based on the updated understandings of redox state-dependent gene regulation in AD are proposed to overcome the lack of efficacy of antioxidant therapies.
Collapse
|
16
|
Liu Y, Lu J, Shi J, Hou Y, Zhu H, Zhao S, Liu H, Ding B, Yin Y, Yi G. Increased expression of the peroxisome proliferator-activated receptor gamma in the immune system of weaned pigs after Escherichia coli lipopolysaccharide injection. Vet Immunol Immunopathol 2008; 124:82-92. [PMID: 18394717 DOI: 10.1016/j.vetimm.2008.02.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2007] [Revised: 02/19/2008] [Accepted: 02/20/2008] [Indexed: 11/30/2022]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARgamma), a member of the nuclear hormone receptor superfamily, has been implicated in regulation of immunity and inflammation in rodents and humans. The objective of the current study was to investigate whether the expression of PPARgamma was altered in the immune system of weaned pigs after Escherichia coli lipopolysaccharide (LPS) injection. PPARgamma expression was investigated in the thymus, spleen, mesenteric lymph node and peripheral white blood cells of weaned pigs (8.54+/-0.24 kg BW) after LPS injection (100 microg/kg BW, n=6) and controls (sterile saline, n=6), by using real-time polymerase chain reaction, Western blot analysis, and immunohistochemistry. Plasma pro-inflammatory cytokines and hormones were also assessed. LPS triggered PPARgamma mRNA and protein expression in the thymus (P<0.05, 4.24-fold; P<0.10, 1.46-fold), spleen (P<0.10, 2.75-fold; P<0.05, 1.84-fold), mesenteric lymph node (P<0.05, 4.32-fold; P<0.05, 1.96-fold) and peripheral white blood cells (P<0.001, 24.44-fold; P<0.001, 1.58-fold). The LPS-injected pigs showed an increase in PPARgamma staining in splenic corpuscle and periarterial lymphatic sheath of white pulp (P<0.05) and red pulp (P<0.001) of spleen, and in medullas of thymus lobule of thymus (P<0.05), and in thymus-dependent area of mesenteric lymph node (P<0.05) compared to the control pigs. Concurrent with up-regulation of PPARgamma expression, LPS induced increases in plasma interleukin-6 (P<0.001), tumor necrosis factor-alpha (P<0.001), cortisol (P<0.001), prostaglandin E(2) (P<0.01) and 15-deoxy-Delta(12,14)-prostaglandin J(2) (15 d-PGJ(2)) (P<0.05), and decreases in plasma insulin (P<0.10) and insulin-like growth factor-1 (P<0.001). These results suggest that induction of PPARgamma expression in immune system may be associated with the release of the natural PPARgamma activating ligand 15 d-PGJ(2), and play an important role in host response to immunological stress. Additionally, it is possible that PPARgamma would be a new therapeutic target in treatment of immunological stress of livestock.
Collapse
Affiliation(s)
- Yulan Liu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan 430023, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Fernyhough ME, Okine E, Hausman G, Vierck JL, Dodson MV. PPARgamma and GLUT-4 expression as developmental regulators/markers for preadipocyte differentiation into an adipocyte. Domest Anim Endocrinol 2007; 33:367-78. [PMID: 17560753 DOI: 10.1016/j.domaniend.2007.05.001] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2007] [Revised: 05/07/2007] [Accepted: 05/08/2007] [Indexed: 12/25/2022]
Abstract
In this document, we have integrated knowledge about two major cellular markers found in cells of the adipocyte lineage (an adipogenic marker and a metabolic marker). This review provides information as to how differentiation of a cell (such as an adipofibroblast, fibroblast or preadipocyte) to become a viable (and new) adipocyte is under different regulation than that experienced by an immature adipocyte that is just beginning to accumulate lipid. The differentiation, prior to lipid-filling, involves PPARgamma. Subsequently, lipid-filling of the adipocyte relies on a late subset of genes and, depending on depot specificity, involves GLUT-4 or any number of other metabolic markers.
Collapse
Affiliation(s)
- M E Fernyhough
- Department of Animal Sciences, Washington State University, PO Box 646310, Pullman, WA 99164, United States
| | | | | | | | | |
Collapse
|
18
|
Qin S, Liu T, Kamanna VS, Kashyap ML. Pioglitazone stimulates apolipoprotein A-I production without affecting HDL removal in HepG2 cells: involvement of PPAR-alpha. Arterioscler Thromb Vasc Biol 2007; 27:2428-34. [PMID: 17872455 DOI: 10.1161/atvbaha.107.150193] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Pioglitazone, an antihyperglycemic drug, increases plasma high-density lipoprotein (HDL)-cholesterol in patients with type 2 diabetes. The mechanisms by which pioglitazone regulate HDL levels are not clear. This study examined the effect of pioglitazone on hepatocyte apolipoprotein AI (apoA-I) and apoA-II production and HDL-protein/cholesterol ester uptake. METHODS AND RESULTS In human hepatoblastoma (HepG2) cells, pioglitazone, dose-dependently (0.5 to 10 micromol/L), increased the de novo synthesis (up to 45%), secretion (up to 44%), and mRNA expression (up to 59%) of apoA-I. Pioglitazone also increased apoA-II de novo synthesis (up to 73%) and mRNA expression (up to 129%). Pioglitazone did not affect the uptake of HDL3-protein or HDL3-cholesterol ester in HepG2 cells. The pioglitazone-induced apoA-I lipoprotein particles increased cholesterol efflux from THP-1 macrophages. The pioglitazone-induced apoA-I secretion or mRNA expression by the HepG2 cells was abrogated with the suppression of PPAR-alpha by small interfering RNA or a specific inhibitor of PPAR-alpha, MK886. CONCLUSIONS The data indicate that pioglitazone increases HDL by stimulating the de novo hepatic synthesis of apoA-I without affecting hepatic HDL-protein or HDL-cholesterol removal. We suggest that pioglitazone-mediated hepatic activation of PPAR-alpha may be one of the mechanisms of action of pioglitazone to raise hepatic apoA-I and HDL.
Collapse
Affiliation(s)
- Shucun Qin
- Atherosclerosis Research Center, Department of Veterans Affairs Healthcare System, Long Beach, California 90822, USA
| | | | | | | |
Collapse
|
19
|
Abstract
Obesity results in marked alterations in cardiac energy metabolism, with a prominent effect being an increase in fatty acid uptake and oxidation by the heart. Obesity also results in dramatic changes in the release of adipokines, such as leptin and adiponectin, both of which have emerged as important regulators of cardiac energy metabolism. The link among obesity, cardiovascular disease, lipid metabolism, and adipokine signaling is complex and not well understood. However, optimizing cardiac energy metabolism in obese subjects may be one approach to preventing and treating cardiac dysfunction that can develop in this population. This review discusses what is presently known about the effects of obesity and the impact adipokines have on cardiac energy metabolism and insulin signaling. The clinical implications of obesity and energy metabolism on cardiac disease are also discussed.
Collapse
Affiliation(s)
- Gary D Lopaschuk
- Cardiovascular Research Group, University of Alberta, Edmonton, Alberta, Canada.
| | | | | |
Collapse
|
20
|
Cabrero A, Cubero M, Llaverías G, Alegret M, Sánchez R, Laguna JC, Vázquez-Carrera M. Leptin down-regulates peroxisome proliferator-activated receptor gamma (PPAR-gamma) mRNA levels in primary human monocyte-derived macrophages. Mol Cell Biochem 2006; 275:173-9. [PMID: 16335797 DOI: 10.1007/s11010-005-1353-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Increased leptin levels are associated with cardiovascular disease in obesity although the mechanism is unknown. Peroxisome proliferator-activated receptor gamma (PPARgamma) is a key regulator of macrophage lipid metabolism and its activation by thiazolidinediones protects against atherosclerosis. The aim of this study was to assess the effects of human recombinant leptin on PPARgamma mRNA levels in primary human macrophages and macrophage-derived foam cells. Leptin treatment (100 ng/ml) for 24 h caused a 41% reduction (p < 0.01) in PPARgamma transcript levels in human-derived macrophages. This fall was accompanied by a reduction in the mRNA expression of carnitine palmitoyltransferase (CPT-I) (36%, p < 0.05) and ABCA1 (62%, p < 0.05), whereas CD36 mRNA reduction (34%) was not significant. In macrophage-derived foam cells, leptin at 20 ng/ml reduced PPARgamma mRNA levels by 33% (p < 0.01) and CPT-I by 27% (p < 0.05). At this concentration, leptin did not modify the expression of either ABCA1 or CD36. In agreement with these results, intracellular cholesterol ester accumulation was not altered in macrophage-derived foam cells by leptin at 20 ng/ml. We propose that the reduction in PPARgamma expression in both macrophages and foam cells may be one of the factors linking high leptin levels and cardiovascular disease.
Collapse
Affiliation(s)
- Agatha Cabrero
- Unitat de Farmacologia, Departament de Farmacologia i Química Terapèutica, Facultat de Farmàcia, University of Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
21
|
Wang K, Zhou Z, Zhang M, Fan L, Forudi F, Zhou X, Qu W, Lincoff AM, Schmidt AM, Topol EJ, Penn MS. Peroxisome Proliferator-Activated Receptor γ Down-Regulates Receptor for Advanced Glycation End Products and Inhibits Smooth Muscle Cell Proliferation in a Diabetic and Nondiabetic Rat Carotid Artery Injury Model. J Pharmacol Exp Ther 2005; 317:37-43. [PMID: 16368901 DOI: 10.1124/jpet.105.095125] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Diabetes is associated with an increase in circulating advanced glycosylation end products (AGEs) and the increased expression of the receptor for AGEs (RAGE). Inhibition of AGE/RAGE binding through the administration of soluble RAGE (sRAGE) has been shown to decrease neointimal hyperplasia. Peroxisome proliferator-activated receptor gamma (PPARgamma), which inhibits neointimal hyperplasia, has been shown to decrease RAGE expression in cultured endothelial cells. We hypothesized that PPARgamma agonists inhibit neointimal hyperplasia via down-regulation of RAGE in vivo. Pretreatment of rat aortic smooth muscle cells (SMCs) with PPARgamma agonist rosiglitazone significantly down-regulated RAGE expression and inhibited SMC proliferation in response to the RAGE agonist S100/calgranulins. In vivo studies showed that rosiglitazone decreased RAGE expression and SMC proliferation at 7 days following carotid arterial injury in both diabetic and nondiabetic rats. At 21 days following injury, neointimal formation was significantly decreased in both diabetic and nondiabetic animals that received rosiglitazone. To determine whether inhibition of neointimal formation by PPARgamma activation could fully be accounted for by its down-regulation of RAGE, we compared the results obtained in animals treated with sRAGE, PPARgamma activator, and sRAGE + PPARgamma activator. Consistent with PPARgamma working through its effects on RAGE, we found that the addition of PPARgamma activator to sRAGE did not result in any further decrease in neointimal formation. These data demonstrate for the first time that PPARgamma agonists inhibit RAGE expression at sites of arterial injury and suggest that down-regulation of RAGE by the PPARgamma activation inhibits neointimal formation in response to arterial injury.
Collapse
Affiliation(s)
- Kai Wang
- Department of Cardiovascular Medicine, Cleveland Clinic Foundation, OH 44195, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cho MC, Lee WS, Hong JT, Park SW, Moon DC, Paik SG, Yoon DY. 5-(3,5-Di-tert-butyl-4-hydroxybenzylidene) thiazolidine-2,4-dione modulates peroxisome proliferators-activated receptor gamma in 3T3-L1 adipocytes: roles as a PPARgamma ligand. Mol Cell Endocrinol 2005; 242:96-102. [PMID: 16171942 DOI: 10.1016/j.mce.2005.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2005] [Accepted: 08/04/2005] [Indexed: 01/01/2023]
Abstract
Based on the binding between peroxisome proliferators-activated receptor gamma (PPARgamma) and steroid receptor co-activator-1 (SRC-1), an enzyme-linked immunosorbent assay (ELISA) was used to screen new PPARgamma ligands from various benzylidinethiazole derivatives, which have anti-inflammatory activity. Among those derivatives, 5-(3,5-di-tert-butyl-4-hydroxybenzylidene) thiazolidine-2,4-dione (BTZD) increased the binding between PPARgamma and SRC-1. BTZD was found to induce adipogenesis and PPARgamma trans-activation in 3T3-L1 pre-adipocyte, and increased the binding between PPARgamma and SRC-1 in in vitro binding assay and complex consisting of PPARgamma and SRC-1 in the co-immunoprecipitaion. Chromatin immunoprecipitation (ChIP) analysis revealed that BTZD induced the binding of PPARgamma-SRC-1 complex to PPAR response element (PPRE) in the same pattern of other PPARgamma ligand. From these studies, we have identified and studied the function of a new PPARgamma ligand, BTZD. We suggest that BTZD can be used as a modulator of PPARgamma. This study applying ELISA and ChIP assay can offer new methods to screen PPARgamma ligand and understand the effects of PPARgamma ligands on inflammation.
Collapse
Affiliation(s)
- Min-Chul Cho
- Laboratory of Cell Biology, Korea Research Institute of Bioscience and Biotechnology, P.O. Box 115, Daejeon 305-600, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
African Americans and other ethnic minority groups suffer disproportionately from type 2 diabetes and its complications than do white Americans. Genetic and environmental factors contribute to the ethnic disparities in diabetes and its complications. The key elements of a comprehensive diabetes management strategy include monitoring, education, dietary modification, exercise, and medications. The progressive nature of diabetes requires the use of more than one agent. Drug combinations should be selected for their therapeutic fire power and complementary mechanisms of action, and exogenous insulin need not be delayed unnecessarily if oral agents are ineffective.
Collapse
Affiliation(s)
- Leonard E Egede
- Division of General Internal Medicine, Department of Medicine, Medical University of South Carolina, Charleston, SC 29425, USA
| | | |
Collapse
|
24
|
Ng TWK, Watts GF, Farvid MS, Chan DC, Barrett PHR. Adipocytokines and VLDL metabolism: independent regulatory effects of adiponectin, insulin resistance, and fat compartments on VLDL apolipoprotein B-100 kinetics? Diabetes 2005; 54:795-802. [PMID: 15734858 DOI: 10.2337/diabetes.54.3.795] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We investigated the relationship of plasma adipocytokine concentrations with VLDL apolipoprotein B (apoB)-100 kinetics in men. Plasma adiponectin, leptin, resistin, interleukin-6 (IL-6), and tumor necrosis factor-alpha (TNF-alpha) concentrations were measured using enzyme immunoassays and insulin resistance by homeostasis model assessment (HOMA) score in 41 men with BMI of 22-35 kg/m(2). VLDL apoB kinetics were determined using an intravenous infusion of 1-[(13)C]leucine, gas chromatography-mass spectrometry, and compartmental modeling. Visceral and subcutaneous adipose tissue mass (ATM) were determined using magnetic resonance imaging, and total ATM was measured by bioelectrical impedance. In univariate regression, plasma adiponectin and leptin concentrations were inversely and directly associated, respectively, with plasma triglyceride; HOMA score; and visceral, subcutaneous, and total ATMs. Conversely, adiponectin and leptin were directly and inversely correlated, respectively, with VLDL apoB catabolism and HDL cholesterol concentration (P < 0.05). Resistin, IL-6, and TNF-alpha were not significantly associated with any of these variables. In multivariate regression, adiponectin was the most significant predictor of plasma VLDL apoB concentration (P = 0.001) and, together with total or subcutaneous ATM, was an independent predictor of VLDL apoB catabolism (P < 0.001); HOMA score was the most significant predictor of VLDL apoB hepatic secretion (P < 0.05). Leptin was not an independent predictor of VLDL apoB kinetics. In conclusion, plasma VLDL apoB kinetics may be differentially controlled by adiponectin and insulin resistance, with adiponectin regulating catabolism and insulin resistance regulating hepatic secretion in men. Total body fat may also independently determine the rate of VLDL catabolism, but leptin, resistin, IL-6, and TNF-alpha do not have a significant effect in regulating apoB kinetics.
Collapse
Affiliation(s)
- Theodore W K Ng
- School of Medicine and Pharmacology, Western Australian Institute for Medical Research, University of Western Australia, Perth, Western Australia 6847, Australia
| | | | | | | | | |
Collapse
|
25
|
Seki N, Bujo H, Jiang M, Shibasaki M, Takahashi K, Hashimoto N, Saito Y. A potent activator of PPARα and γ reduces the vascular cell recruitment and inhibits the intimal thickning in hypercholesterolemic rabbits. Atherosclerosis 2005; 178:1-7. [PMID: 15585194 DOI: 10.1016/j.atherosclerosis.2004.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2004] [Revised: 07/18/2004] [Accepted: 08/10/2004] [Indexed: 11/28/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) regulate the vascular cell functions as well as systemic lipid and glucose metabolism. Here, we studied the effect of TAK-559, a newly developed potent activator both for PPARalpha and gamma, on the vascular cell recruitment. TNF-alpha- or interleukin-1beta (IL-1beta)-induced THP-1 cell attachment to cultured endothelial cells was significantly reduced in the presence of 10 microM TAK-559 (P < 0.05). The secretion of monocyte chemoattractant protein-1 (MCP-1) from endothelial cells is reduced by 36% in the presence of 10 microM TAK-559, accompanied with the decreased mRNA expression in the cells. The proliferation and migration of cultured smooth muscle cells (SMCs) were significantly decreased in the presence of TAK-559 (P < 0.05). TAK-559-treated hypercholesterolemic rabbits showed the significant reduction of intimal thickning after balloon catheterization by 51% compared with control (P < 0.05), although the plasma lipid and glucose level was not changed between them. The numbers of macrophage and SMCs were decreased to 34% and 49% in the hyperplastic intima of arteries from TAK-559-treated rabbits compared to those from control, respectively. These results suggest that the PPARalpha and gamma activator inhibits the recruitment of macrophages and SMCs in intima, possibly leading to the reduction of intimal hyperplasia in hypercholesterolemia.
Collapse
Affiliation(s)
- Naoto Seki
- Department of Clinical Cell Biology (F5), Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | | | | | | | | | | | | |
Collapse
|
26
|
Herrera-Arellano A, Aguilar-Santamaría L, García-Hernández B, Nicasio-Torres P, Tortoriello J. Clinical trial of Cecropia obtusifolia and Marrubium vulgare leaf extracts on blood glucose and serum lipids in type 2 diabetics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2004; 11:561-566. [PMID: 15636168 DOI: 10.1016/j.phymed.2004.01.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Cecropia obtusifolia and Marrubium vulgare have been widely used in Mexican traditional medicine for the control of type 2 diabetes. In order to evaluate the clinical effect produced by the aqueous extract from these species on type 2 non-controlled diabetes mellitus, a total of 43 outpatients were included. Based on the European NIDDM (policy group) criteria, only patients with poor response to the conventional treatment were selected. All patients maintained their medical treatment and also received a prepared infusion of the dry leaves of the plant treatment for 21 days. In a double-blind manner, the patients were randomly grouped as follows: 22 patients were treated with C. obtusifolia and 21 with M. vulgare. The fasting blood glucose values were reduced by 15.25% on patients treated with C. obtusifolia, while cholesterol and triglycerides were decreased by 14.62% and 42.0%, respectively (ANOVA p< 0.02). In the case of patients treated with M. vulgare, the plasma glucose level was reduced by 0.64% and cholesterol and triglycerides by 4.16% and 5.78%, respectively. When the results were compared between groups, significant differences in glucose and cholesterol diminution were found. The obtained results showed that the infusion prepared with the leaves of C. obtusifolia (containing 2.99+/-0.14mg of chlorogenic acid/g of dried plant) produced beneficial effects on carbohydrate and lipid metabolisms when it was administered as an adjunct on patients with type 2 diabetes with poor response to conventional medical treatment.
Collapse
Affiliation(s)
- A Herrera-Arellano
- Centro de Investigación Biomédica del Sur, Instituto Mexicano del Seguro Social, Xochitepec, Morelos, México.
| | | | | | | | | |
Collapse
|
27
|
Abstract
Cardiovascular disease is significantly increased in patients with metabolic syndrome and type 2 diabetes. Several factors such as chronic hyperglycemia, lipId abnormalities, endothelium dysfunction, inflammation, oxIdative stress, increased thrombosis and decreased fibrinolysis are likely to promote cardiovascular events in these patients. Because of positive effects on glucose homeostasis, lipId metabolism, proteins involved in all stages of atherogenesis, endothelium function, inflammation, thrombosis and fibrinolysis, PPARS alpha (fibrates) and PPARs gamma (glitazones) agonists are good candIdates to reduce cardiovascular disease, more precisely in subjects with metabolic syndrome or type 2 diabetes. PPARS alpha agonists (fibrates) are potent hypolipIdemic agents increasing plasma HDL-cholesterol and reducing free fatty acIds, triglycerIdes, LDL-cholesterol and the number of small dense LDL pArticles. Moreover, they reduce vascular inflammation and thrombosis, promote fibrinolysis and inhibit the production of the vasoconstrictor factor, endothelin-1, by the endothelium. They have been shown, in clinical trials, to reduce cardiovascular disease, more particularly in patients displaying lipId abnormalities typical of metabolic syndrome and type 2 diabetes (high triglycerIdes, low HDL-cholesterol). PPARS gamma agonists (glitazones) have not only beneficial effects on glucose homeostasis, by increasing insulin sensitivity and reducing blood glucose level but also on lipId metabolism by elevating plasma HDL-cholesterol, decreasing free fatty acIds and the number of small dense LDL pArticles, and for pioglitazone by reducing plasma triglycerIdes. Furthermore, they diminish vascular inflammation and vasoconstriction, inhibit monocyte chemotaxis, proliferation and migration of smooth muscle cells, in the vascular wall and decrease the production of adhesion molecules and metalloproteinases. PPARs gamma agonists (glitazones) have been shown to reduce the development of atherosclerotic lesions in rats. The potential clinical benefit of PPARs gamma agonists on the reduction of cardiovascular disease, in type 2 diabetic patients, will be specified by the ongoing intervention studies.
Collapse
Affiliation(s)
- B Vergès
- Service d'Endocrinologie, Diabétologie et Maladies Métaboliques, hôpital du Bocage, CHU de Dijon, France.
| |
Collapse
|
28
|
Abstract
Atherosclerosis is an inflammatory disease of the vessel wall, characterized by the accumulation of leukocytes, especially macrophages and T-cells. Chemokines are small heparin-binding polypeptides, whose main function is to attract cells to the areas of developing inflammation. They function by ligating G-protein coupled chemokine receptors initiating different signaling cascades. In vivo and in vitro investigations showed that chemokines are produced by a variety of cells and play important roles in the development and progression of many physiological and pathological conditions including atherosclerosis. Chemokines such as MCP-1, MCP-4, MIP-1 and RANTES may mediate leukocyte trafficking to, and their retention in, the plaque while CXCL16 seems to fulfill the dual function of a chemokine and a scavenger receptor. Chemokine and chemokine receptor homologues are secreted by several viruses, which may also play a role in the pathogenesis of atherosclerosis. Expression levels and gene polymorphisms of some chemokines may become useful clinical markers of atherosclerosis and other cardiovascular diseases. Modulation of chemokines and chemokine receptors' expression as well as their signaling pathways may provide important anti-atherogenic strategies.
Collapse
Affiliation(s)
- Yuri Sheikine
- Center for Molecular Medicine, Cardiovascular Research Unit, Karolinska Institute, Stockholm, Sweden.
| | | |
Collapse
|
29
|
Bonfield TL, Farver CF, Barna BP, Malur A, Abraham S, Raychaudhuri B, Kavuru MS, Thomassen MJ. Peroxisome proliferator-activated receptor-gamma is deficient in alveolar macrophages from patients with alveolar proteinosis. Am J Respir Cell Mol Biol 2003; 29:677-82. [PMID: 12805087 DOI: 10.1165/rcmb.2003-0148oc] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) is a ligand-activated, nuclear transcription factor that regulates genes involved in lipid and glucose metabolism, inflammation, and other pathways. The hematopoietic growth factor, granulocyte macrophage colony-stimulating factor (GM-CSF), is essential for lung homeostasis and is thought to regulate surfactant clearance, but mechanisms involved are unknown. GM-CSF is reported to stimulate PPAR-gamma, but the activation status of PPAR-gamma in human alveolar macrophages has not been defined. In pulmonary alveolar proteinosis (PAP), a rare interstitial lung disease, surfactant accumulates in alveolar airspaces, resident macrophages become engorged with lipoproteinaceous material, and GM-CSF deficiency is strongly implicated in pathogenesis. Here we show that PPAR-gamma mRNA and protein are highly expressed in alveolar macrophages of healthy control subjects but severely deficient in PAP in a cell-specific manner. Further, we show that the PPAR-gamma-regulated lipid scavenger receptor, CD36, is also deficient in PAP. PPAR-gamma and CD36 deficiency are not intrinsic to PAP alveolar macrophages, but can be upregulated by GM-CSF therapy. Moreover, GM-CSF treatment of patients with PAP fully restores PPAR-gamma to healthy control levels. Based upon these novel findings, we hypothesize that GM-CSF regulates lung homeostasis via PPAR-gamma-dependent pathways.
Collapse
Affiliation(s)
- Tracey L Bonfield
- Department of Pulmonary and Critical Care Medicine, 9500 Euclid Avenue, Cleveland Clinic Foundation, Desk A90, Cleveland, OH 44195-5038.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Mehta JL, Hu B, Chen J, Li D. Pioglitazone inhibits LOX-1 expression in human coronary artery endothelial cells by reducing intracellular superoxide radical generation. Arterioscler Thromb Vasc Biol 2003; 23:2203-8. [PMID: 12958047 DOI: 10.1161/01.atv.0000094411.98127.5f] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE LOX-1, a novel lectin-like receptor for oxidized LDL (ox-LDL), is expressed in response to ox-LDL, angiotensin II (Ang II), tumor necrosis factor (TNF)-alpha, and other stress stimuli. It is highly expressed in atherosclerotic tissues. Peroxisome proliferator-activated receptor (PPAR)-gamma ligands, such as pioglitazone, exert antiatherosclerotic effects. This study examined the regulation of LOX-1 expression in human coronary artery endothelial cells (HCAECs) by pioglitazone. METHODS AND RESULTS Fourth generation HCAECs were treated with ox-LDL, Ang II, or TNF-alpha with or without pioglitazone pretreatment. All 3 stimuli upregulated LOX-1 expression (mRNA and protein). Pioglitazone, in a concentration-dependent manner, reduced LOX-1 expression (P<0.01 versus ox-LDL, Ang II, or TNF-alpha alone). Ox-LDL, Ang II, and TNF-alpha each enhanced intracellular superoxide radical generation, and pioglitazone pretreatment reduced superoxide generation (P<0.01 versus ox-LDL, Ang II, or TNF-alpha). Furthermore, all 3 stimuli upregulated the expression of the transcription factors nuclear factor-kappaB and activator protein-1 (determined by electrophoretic mobility shift assay), and pioglitazone pretreatment reduced this expression (P<0.01 versus ox-LDL, Ang II, or TNF-alpha). To determine the biological significance of pioglitazone-mediated downregulation of LOX-1, we studied monocyte adhesion to ox-LDL-treated HCAECs. Pioglitazone reduced the adhesion of monocytes to activated HCAECs in a fashion similar to that produced by antisense to LOX-1 mRNA. CONCLUSIONS These observations suggest that the PPAR-gamma ligand pioglitazone reduces intracellular superoxide radical generation and subsequently reduces the expression of transcription factors, expression of the LOX-1 gene, and monocyte adhesion to activated endothelium. The salutary effect of PPAR-gamma ligands in atherogenesis may involve the inhibition of LOX-1 and the adhesion of monocytes to endothelium.
Collapse
Affiliation(s)
- Jawahar L Mehta
- Division of Cardiovascular Medicine, University of Arkansas for Medical Sciences and Central Arkansas Veterans Healthcare System, 4301 W Markham St, No. 532, Little Rock, AR 72205, USA.
| | | | | | | |
Collapse
|
31
|
Abstract
This review focuses on the role of monocytes in the early phase of atherogenesis, before foam cell formation. An emerging consensus underscores the importance of the cellular inflammatory system in atherogenesis. Initiation of the process apparently hinges on accumulating low-density lipoproteins (LDL) undergoing oxidation and glycation, providing stimuli for the release of monocyte attracting chemokines and for the upregulation of endothelial adhesive molecules. These conditions favor monocyte transmigration to the intima, where chemically modified, aggregated, or proteoglycan- or antibody-complexed LDL may be endocytotically internalized via scavenger receptors present on the emergent macrophage surface. The differentiating monocytes in concert with T lymphocytes exert a modulating effect on lipoproteins. These events propagate a series of reactions entailing generation of lipid peroxides and expression of chemokines, adhesion molecules, cytokines, and growth factors, thereby sustaining an ongoing inflammatory process leading ultimately to lesion formation. New data emerging from studies using transgenic animals, notably mice, have provided novel insights into many of the cellular interactions and signaling mechanisms involving monocytes/macrophages in the atherogenic processes. A number of these studies, focusing on mechanisms for monocyte activation and the roles of adhesive molecules, chemokines, cytokines and growth factors, are addressed in this review.
Collapse
Affiliation(s)
- Bjarne Osterud
- Department of Biochemistry, Institute of Medical Biology, Faculty of Medicine, University of Tromsø, Tromsø, Norway.
| | | |
Collapse
|
32
|
Culver DA, Barna BP, Raychaudhuri B, Bonfield TL, Abraham S, Malur A, Farver CF, Kavuru MS, Thomassen MJ. Peroxisome proliferator-activated receptor gamma activity is deficient in alveolar macrophages in pulmonary sarcoidosis. Am J Respir Cell Mol Biol 2003; 30:1-5. [PMID: 14512375 DOI: 10.1165/rcmb.2003-0304rc] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The ligand-activated transcription factor, peroxisome proliferator-activated receptor gamma (PPAR gamma), has pleiotropic effects on lipid and glucose metabolism as well as modulating immune activity. In Th1-predominant models of inflammatory bowel disease and arthritis, PPAR gamma ligands can ameliorate clinical disease severity, partly by downregulating a range of inflammatory cytokines. However, PPAR gamma has not been evaluated in chronic sarcoidosis, a disease characterized by persistent activation of Th1 immune responses in alveolar macrophages. We hypothesized that a deficiency of PPAR gamma activity contributes to ongoing inflammation in pulmonary sarcoidosis via failure to repress proinflammatory transcription factors. To address this, we studied eight patients with active sarcoidosis and nine healthy control subjects by bronchoscopy. Bronchoalveolar lavage specimens from patients revealed a striking reduction of PPAR gamma activity by electrophoretic mobility shift assay in alveolar macrophages compared with healthy control subjects, with a concomitant upregulation of nuclear factor (NF)-kappa B activity. Immunostaining and real-time polymerase chain reaction demonstrated reductions of PPAR gamma nuclear protein and gene expression. The data show for the first time that alveolar macrophages from patients with active sarcoidosis exhibit activation of NF-kappa B and deficiency of PPAR gamma. Although these results do not demonstrate a direct causal effect, they are consistent with the hypothesis that insufficient PPAR gamma activity contributes to ongoing dysregulated inflammation in pulmonary sarcoidosis by failing to suppress NF-kappa B.
Collapse
Affiliation(s)
- Daniel A Culver
- Department of Pulmonary and Critical Care Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Dagogo-Jack S. Ethnic disparities in type 2 diabetes: pathophysiology and implications for prevention and management. J Natl Med Assoc 2003; 95:774, 779-89. [PMID: 14527045 PMCID: PMC2594463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Abstract
Based on 2000 census data, ethnic minorities constitute approximately 25% of the overall population of the United States, and the population of minority groups has been increasing at a faster rate than the general U.S. population. Compared with caucasians, persons from minority ethnic groups suffer disproportionately from type 2 diabetes and its long-term complications. Acute complications of diabetes occur with varying frequencies in the different demographic groups, but there are indications that the rate of hospitalization for diabetic ketoacidosis and nonketotic coma may be higher among certain minority populations. Genetic and lifestyle factors likely account for the increased prevalence of type 2 diabetes among ethnic minorities. However, the increase in morbidity and mortality from diabetes may be the result, in part, of socioeconomic factors. Pathophysiologically, several studies have documented a higher prevalence of insulin resistance in minority groups, even after correction for obesity and lifestyle factors. These findings underscore the need for a more aggressive approach to diabetes management in high-risk populations. Behavioral and pharmacologic interventions that reduce insulin resistance have profound beneficial effects in African-American patients and subjects with diabetes from other ethnic groups. Indeed, much of the ethnic difference in morbidity from diabetic complications disappears when caucasians and non-caucasians are treated to identical degrees of glycemic control.
Collapse
Affiliation(s)
- Samuel Dagogo-Jack
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Tennessee Health Sciences Center, 951 Court Avenue, Room 335M, Memphis, TN 38163, USA.
| |
Collapse
|
34
|
Abstract
Diabetes, obesity, atherosclerosis and cancer are the principal contributors to morbidity and mortality in Western society. Emerging evidence indicates that a nuclear receptor, the peroxisome proliferator-activated receptor gamma (PPARgamma), plays a role in these pathological processes. Furthermore, modulation of receptor action in these diseases may be of therapeutic value, as exemplified by the recent introduction of the thiazolidinediones, a novel class of insulin-sensitizing agent for the treatment of type 2 diabetes mellitus. The availability of such high-affinity ligands has facilitated the study of signalling pathways through which PPARgamma regulates metabolic processes; these analyses have been complemented by the study of human subjects harbouring (naturally occurring) mutations and polymorphisms within the receptor. The latter have provided unique genetic evidence for a link between PPARgamma and mammalian glucose homeostasis, lipid metabolism and regulation of fat mass. This review highlights recent studies which have advanced our understanding of the pivotal role that this receptor plays in metabolism, with particular reference to the consequences of inherited variation in the human receptor gene.
Collapse
Affiliation(s)
- Mark Gurnell
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK.
| |
Collapse
|
35
|
Takahashi N, Kawada T, Goto T, Kim CS, Taimatsu A, Egawa K, Yamamoto T, Jisaka M, Nishimura K, Yokota K, Yu R, Fushiki T. Abietic acid activates peroxisome proliferator-activated receptor-gamma (PPARgamma) in RAW264.7 macrophages and 3T3-L1 adipocytes to regulate gene expression involved in inflammation and lipid metabolism. FEBS Lett 2003; 550:190-4. [PMID: 12935909 DOI: 10.1016/s0014-5793(03)00859-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Abietic acid is one of the terpenoids, which are multifunctional natural compounds. It has been reported that abietic acid suppresses effects on inflammation. However, the mechanism underlying the anti-inflammatory effects remains unclear. The present work indicates that abietic acid suppresses the protein expression of tumor necrosis factor-alpha and cyclooxygenase 2, which are involved in inflammation, in lipopolysaccharide-stimulated macrophages. Moreover, this effect resembles that of thiazolidinedione, a synthetic peroxisome proliferator-activated receptor-gamma (PPARgamma) ligand. Indeed, abietic acid activates PPARgamma in luciferase reporter assays. The activity of abietic acid induces PPARgamma target gene expression in RAW264.7 macrophages and 3T3-L1 adipocytes. These data indicate that abietic acid is a PPARgamma ligand and that its anti-inflammatory effect is partly due to the activation of PPARgamma in stimulated macrophages. The present work suggests a novel possibility that abietic acid, a naturally occurring compound, can be used not only for anti-inflammation but also for regulating lipid metabolism and atherosclerosis.
Collapse
Affiliation(s)
- Nobuyuki Takahashi
- Laboratory of Nutrition Chemistry, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Puddu P, Puddu GM, Muscari A. Peroxisome proliferator-activated receptors: are they involved in atherosclerosis progression? Int J Cardiol 2003; 90:133-40. [PMID: 12957742 DOI: 10.1016/s0167-5273(02)00565-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Peroxisome proliferator-activated receptors (PPAR) are nuclear receptors present in several organs and cell types. They are subdivided into PPAR alpha, PPAR gamma and PPAR delta (or beta). PPAR alpha and gamma are the two main categories of these receptors, which are both characterized by their ability to influence lipid metabolism, glucose homeostasis, cell proliferation, differentiation and apoptosis, as well as the inflammatory response, by transcriptional activation of target genes. PPAR alpha are activated by fatty acids, eicosanoids and fibrates, while PPAR gamma activators include arachidonic acid metabolites, oxidized low density lipoprotein and thiazolidinediones. Atherosclerosis is now considered a chronic inflammatory condition. Thus, PPAR activation appears a promising approach to favorably affect atherosclerosis development through both metabolic and anti-inflammatory effects. However, the clinical data in favor of an anti-atherosclerotic action of PPAR agonists are still scanty, and some experimental data would even indicate possible pro-atherogenic effects, or a lack of effect in the female sex. New controlled clinical studies will provide the information necessary to understand the true significance and usefulness of PPAR alpha, gamma and delta activators in the control of atherosclerotic disease.
Collapse
Affiliation(s)
- Paolo Puddu
- Department of Internal Medicine, University of Bologna, S Orsola Hospital, Via Massarenti 9, 40138, Bologna, Italy
| | | | | |
Collapse
|
37
|
Hurst CH, Waxman DJ. Activation of PPARalpha and PPARgamma by environmental phthalate monoesters. Toxicol Sci 2003; 74:297-308. [PMID: 12805656 DOI: 10.1093/toxsci/kfg145] [Citation(s) in RCA: 365] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Phthalate esters are widely used as plasticizers in the manufacture of products made of polyvinyl chloride. Mono-(2-ethylhexyl)-phthalate (MEHP) induces rodent hepatocarcinogenesis by a mechanism that involves activation of the nuclear transcription factor peroxisome proliferator-activated receptor-alpha (PPARalpha). MEHP also activates PPAR-gamma (PPARgamma), which contributes to adipocyte differentiation and insulin sensitization. Human exposure to other phthalate monoesters, including metabolites of di-n-butyl phthalate and butyl benzyl phthalate, is substantially higher than that of MEHP, prompting this investigation of their potential for PPAR activation, assayed in COS cells and in PPAR-responsive liver (PPARalpha) and adipocyte (PPARgamma) cell lines. Monobenzyl phthalate (MBzP) and mono-sec-butyl phthalate (MBuP) both increased the COS cell transcriptional activity of mouse PPARalpha, with effective concentration for half-maximal response (EC50) values of 21 and 63 microM, respectively. MBzP also activated human PPARalpha (EC50=30 microM) and mouse and human PPARgamma (EC50=75-100 microM). MEHP was a more potent PPAR activator than MBzP or MBuP, with mouse PPARalpha more sensitive to MEHP (EC50=0.6 microM) than human PPARalpha (EC50=3.2 microM). MEHP activation of PPARgamma required somewhat higher concentrations, EC50=10.1 microM (mouse PPARgamma) and 6.2 microM (human PPARgamma). No significant PPAR activation was observed with the monomethyl, mono-n-butyl, dimethyl, or diethyl esters of phthalic acid. PPARalpha activation was verified in FAO rat liver cells stably transfected with PPARalpha, where expression of several endogenous PPARalpha target genes was induced by MBzP, MBuP, and MEHP. Similarly, activation of endogenous PPARgamma target genes was evidenced for all three phthalates by the stimulation of PPARgamma-dependent adipogenesis in the 3T3-L1 cell differentiation model. These findings demonstrate the potential of environmental phthalate monoesters for activation of rodent and human PPARs and may help to elucidate the molecular basis for the adverse health effects proposed to be associated with human phthalate exposure.
Collapse
Affiliation(s)
- Christopher H Hurst
- Department of Biology, Division of Cell and Molecular Biology, Boston University, Boston, Massachusetts 02215, USA
| | | |
Collapse
|
38
|
Abstract
Why inflammatory responses become chronic and how adjuvants work remain unanswered. Macrophage-lineage cells are key components of chronic inflammatory reactions and in the actions of immunologic adjuvants. One explanation for the increased numbers of macrophages long term at sites of chronic inflammation could be enhanced cell survival or even local proliferation. The evidence supporting a unifying hypothesis for one way in which this macrophage survival and proliferation may be promoted is presented. Many materials, often particulate, of which macrophages have difficulty disposing, can promote monocyte/macrophage survival and even proliferation. Materials active in this regard and which can initiate chronic inflammatory reactions include oxidized low-density lipoprotein, inflammatory microcrystals (calcium phosphate, monosodium urate, talc, calcium pyrophosphate), amyloidogenic peptides (amyloid beta and prion protein), and joint implant biomaterials. Additional, similar materials, which have been shown to have adjuvant activity (alum, oil-in-water emulsions, heat-killed bacteria, CpG oligonucleotides, methylated bovine serum albumin, silica), induce similar responses. Cell proliferation can be striking, following uptake of some of the materials, when macrophage-colony stimulating factor is included at low concentrations, which normally promote mainly survival. It is proposed that if such responses were occurring in vivo, there would be a shift in the normal balance between cell survival and cell death, which maintains steady-state, macrophage-lineage numbers in tissues. Thus, there would be more cells in an inflammatory lesion or at a site of adjuvant action with the potential, following activation and/or differentiation, to perpetuate inflammatory or antigen-specific, immune responses, respectively.
Collapse
Affiliation(s)
- John A Hamilton
- Arthritis and Inflammation Research Centre and Cooperative Research Centre for Chronic Inflammatory Diseases, University of Melbourne, Department of Medicine, The Royal Melbourne Hospital, Parkville, Australia.
| |
Collapse
|
39
|
Cabrero A, Cubero M, Llaverías G, Jové M, Planavila A, Alegret M, Sánchez R, Laguna JC, Carrera MV. Differential effects of peroxisome proliferator-activated receptor activators on the mRNA levels of genes involved in lipid metabolism in primary human monocyte-derived macrophages. Metabolism 2003; 52:652-7. [PMID: 12759900 DOI: 10.1053/meta.2003.50100] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Peroxisome proliferator-activated receptors (PPARs) are key regulators of macrophage lipid metabolism. We compared the effects of three PPAR activators (bezafibrate, fenofibrate, and troglitazone) on the mRNA levels of genes involved in lipid metabolism in primary human macrophages and macrophage-derived foam cells. Treatment of human macrophages for 24 hours with 100 micro mol/L bezafibrate, a nonselective drug that activates the 3 PPAR subtypes (PPARalpha, PPARbeta/delta, and PPARgamma), caused an 87% (P <.01) and a 230% rise in CD36 and adipocyte fatty acid-binding protein (aP2) mRNA levels, respectively, whereas the expressions of PPARgamma, PPARalpha, acyl-CoA oxidase, carnitine palmitoyltransferase I (CPT-I), adenosine triphosphate (ATP)-binding cassette transporter 1 (ABCA1), neutral cholesteryl ester hydrolase, and lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) were not modified. However, treatment with selective PPARalpha (fenofibrate at 100 micro mol/L) and PPARgamma (troglitazone at 5 micro mol/L) activators had different effects. Fenofibrate increased PPARalpha (62%, P <.05) and LOX-1 (180%, P <.05) mRNA levels; and troglitazone upregulated CPT-I expression (75%, P <.05). When the effects of these drugs were assessed in macrophage-derived foam cells, we found that troglitazone caused a 134% (P <.05) and a 66% (P <.01) rise in ABCA1 and CPT-I mRNA levels, respectively, whereas the 3 drugs significantly increased aP2 transcripts (about 100% induction). Given that troglitazone treatment resulted in the upregulation of genes involved in the mitochondrial beta-oxidation of fatty acids (CPT-I) and in the reverse-cholesterol-transport pathway (ABCA1), we subsequently determined whether these changes affected intracellular cholesterol ester accumulation. In macrophage-derived foam cells a significant reduction (32%, P <.01) was observed in intracellular cholesterol accumulation after troglitazone, but not after bezafibrate or fenofibrate treatment. Since CPT-I inhibition promotes cholesterol incorporation into cholesteryl esters in macrophages, study is now needed on whether CPT-I induction by troglitazone may reduce the availability of fatty acids for synthesizing cholesterol esters, leading to less foam cell formation.
Collapse
Affiliation(s)
- Agatha Cabrero
- Unitat de Farmacologia, Departament de Farmacologia i Química Terapèutica, Facultat de Farmàcia, University of Barcelona, Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Argmann CA, Sawyez CG, McNeil CJ, Hegele RA, Huff MW. Activation of peroxisome proliferator-activated receptor gamma and retinoid X receptor results in net depletion of cellular cholesteryl esters in macrophages exposed to oxidized lipoproteins. Arterioscler Thromb Vasc Biol 2003; 23:475-82. [PMID: 12615696 DOI: 10.1161/01.atv.0000058860.62870.6e] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Peroxisome proliferator-activated receptor gamma (PPARgamma), a ligand-activated transcription factor, has pleiotropic effects, including regulation of macrophage differentiation and lipid homeostasis. The PPARgamma ligands, thiazolidinediones (TZDs), attenuate atherosclerosis in mice by uncertain mechanisms. The objective of this study was to determine whether activation of PPARgamma or its obligate heterodimer, retinoid X receptor (RXR), modulates macrophage foam cell formation induced by oxidized (ox) lipoproteins. METHODS AND RESULTS Incubation of THP-1 macrophages with oxHTG-VLDL, oxREM, or oxLDL increased cellular cholesteryl ester over 6-fold. Preincubation with the TZD, ciglitazone, the RXR-specific ligand, 9-cis retinoic acid (9cRA) or the combination reduced CE mass accumulation by up to 65%. Ciglitazone and 9cRA increased CD36 mRNA (up to 4-fold); however, uptake of [125I]oxLDL was only modestly enhanced (up to 1.8-fold) becaues of a concomitant PPARgamma:RXR-induced decrease in SRAI/II activity (up to 40%). This suggested that PPARgamma:RXR activation inhibited cholesteryl ester accumulation by enhancing cholesterol efflux. Ciglitazone and 9cRA were found to increase the expression of ATP-binding cassette proteins A1 and G1, resulting in enhanced cholesterol efflux to lipoprotein-deficient serum, apoAI and HDL3. CONCLUSIONS PPARgamma and/or RXR activation inhibit foam cell formation through enhanced cholesterol efflux despite increased oxLDL uptake. These observations explain the reduced atherosclerosis in TZD-treated mice and may extend the therapeutic implications of these ligands.
Collapse
Affiliation(s)
- C A Argmann
- Department of Medicine and Robarts Research Institute, University of Western Ontario, London, Ontario, Canada N6A 5K8
| | | | | | | | | |
Collapse
|
41
|
Kasono K, Nishida J, Tamemoto H, Fudaka K, Namai K, Kajio H, Masatoshi K, Kanazawa Y, Kawakami M. Thiazolidinediones increase the number of platelets in immune thrombocytopenic purpura mice via inhibition of phagocytic activity of the reticulo-endothelial system. Life Sci 2002; 71:2037-52. [PMID: 12175897 DOI: 10.1016/s0024-3205(02)01950-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Thiazolidinediones (TZDs) have broad spectrum of actions, including immunomodulating effects that are dependent or independent of the target nuclear receptor, peroxisome proliferator activated receptor-gamma (PPAR-gamma). In this study, we investigated the effect of TZDs on the platelet numbers in male immune thrombocytopenic purpura (ITP) model mice, (NZW x BXSB)F(1) (W/BF(1)) in vivo, and attempted to clarify the mechanism of action. Seven-day treatment with troglitazone increased platelet counts by 66% compared with those of controls. Within two weeks after the termination of the treatment period, the numbers of platelets were decreased to the level in controls. Pioglitazone showed only weak increasing effect on platelet counts in short-term experiment. However, long-term treatment with the drug resulted in a more pronounced up-regulation of platelets. We next assayed the platelet-associated antibodies (PAA) and the survival rate of antibody-sensitized mouse erythrocytes (Ab-mRBC) in W/BF1 mice. Pioglitazone slightly decreased the production of PAA and significantly elongated the survival period of Ab-mRBC in vivo. These drugs showed dose-dependent inhibitory effects on the cell proliferation and Fcgamma receptor (FcgammaR)-mediated phagocytic activity of macrophage-like cells in vitro. These results suggest that TZDs improve platelet counts in this mouse model mainly by suppressing systemic reticulo-endothelial phagocytic function.
Collapse
MESH Headings
- Animals
- Antibody-Dependent Cell Cytotoxicity/drug effects
- Autoantibodies/analysis
- Blood Platelets/drug effects
- Blood Platelets/immunology
- COS Cells
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Erythrocytes/immunology
- Hybridization, Genetic
- Hypoglycemic Agents/pharmacology
- Macrophages/immunology
- Male
- Mice
- Mice, Inbred Strains
- Mononuclear Phagocyte System/drug effects
- Mononuclear Phagocyte System/immunology
- Phagocytosis/drug effects
- Pioglitazone
- Platelet Count
- Purpura, Thrombocytopenic, Idiopathic/blood
- Purpura, Thrombocytopenic, Idiopathic/drug therapy
- Purpura, Thrombocytopenic, Idiopathic/immunology
- Receptors, Fc/immunology
- Thiazoles/pharmacology
- Thiazolidinediones
Collapse
Affiliation(s)
- Keizo Kasono
- Department of Endocrinology and Metabolism, Jichi Medical School, Omiya Medical Center, 1-847 Amanuma-cho, Saitama 330-8503, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Bernal-Mizrachi C, Weng S, Li B, Nolte LA, Feng C, Coleman T, Holloszy JO, Semenkovich CF. Respiratory uncoupling lowers blood pressure through a leptin-dependent mechanism in genetically obese mice. Arterioscler Thromb Vasc Biol 2002; 22:961-8. [PMID: 12067905 DOI: 10.1161/01.atv.0000019404.65403.71] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Insulin resistance is commonly associated with hypertension, a condition that causes vascular disease in people with obesity and type 2 diabetes. The mechanisms linking hypertension and insulin resistance are poorly understood. To determine whether respiratory uncoupling can prevent insulin resistance-related hypertension, we crossed transgenic mice expressing uncoupling protein 1 (UCP1) in skeletal muscle with lethal yellow (A(y)/a) mice, genetically obese animals known to have elevated blood pressure. Despite increased food intake, UCP-A(y)/a mice weighed less than their A(y)/a littermates. The metabolic rate was higher in UCP-A(y)/a mice than in A(y)/a mice and did not impair their ability to alter oxygen consumption in response to temperature changes, an adaptation involving sympathetic nervous system activity. Compared with their nontransgenic littermates, UCP-A(y)/a mice had lower fasting insulin, glucose, triglyceride, and cholesterol levels and were more insulin sensitive. Blood pressure, serum leptin, and urinary catecholamine levels were also lower in uncoupled mice. Independent of sympathetic nervous system activity, low-dose peripheral leptin infusion increased blood pressure in UCP-A(y)/a mice but not in their A(y)/a littermates. These data indicate that skeletal muscle respiratory uncoupling reverses insulin resistance and lowers blood pressure in genetic obesity without affecting thermoregulation. The data also suggest that uncoupling could decrease the risk of atherosclerosis in type 2 diabetes.
Collapse
Affiliation(s)
- Carlos Bernal-Mizrachi
- Department of Medicine, Washington University School of Medicine, St. Louis, Mo 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Goetze S, Eilers F, Bungenstock A, Kintscher U, Stawowy P, Blaschke F, Graf K, Law RE, Fleck E, Gräfe M. PPAR activators inhibit endothelial cell migration by targeting Akt. Biochem Biophys Res Commun 2002; 293:1431-7. [PMID: 12054675 DOI: 10.1016/s0006-291x(02)00385-6] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Peroxisome proliferator-activated receptors (PPARs) regulate lipid and glucose metabolism and exert several vascular effects that may provide a dual benefit of these receptors on metabolic disorders and atherosclerotic vascular disease. Endothelial cell migration is a key event in the pathogenesis of atherosclerosis. We therefore investigated the effects of lipid-lowering PPARalpha-activators (fenofibrate, WY14643) and antidiabetic PPARgamma-activators (troglitazone, ciglitazone) on this endothelial cell function. Both PPARalpha- and PPARgamma-activators significantly inhibited VEGF-induced migration of human umbilical vein endothelial cells (EC) in a concentration-dependent manner. Chemotactic signaling in EC is known to require activation of two signaling pathways: the phosphatidylinositol-3-kinase (PI3K)-->Akt- and the ERK1/2 mitogen-activated protein kinase (ERK MAPK) pathway. Using the pharmacological PI3K-inhibitor wortmannin and the ERK MAPK-pathway inhibitor PD98059, we observed a complete inhibition of VEGF-induced EC migration. VEGF-induced Akt phosphorylation was significantly inhibited by both PPARalpha- and gamma-activators. In contrast, VEGF-stimulated ERK MAPK-activation was not affected by any of the PPAR-activators, indicating that they inhibit migration either downstream of ERK MAPK or independent from this pathway. These results provide first evidence for the antimigratory effects of PPAR-activators in EC. By inhibiting EC migration PPAR-activators may protect the vasculature from pathological alterations associated with metabolic disorders.
Collapse
Affiliation(s)
- Stephan Goetze
- Department of Medicine/Cardiology, German Heart Institute Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Zhou J, Wilson KM, Medh JD. Genetic analysis of four novel peroxisome proliferator activated receptor-gamma splice variants in monkey macrophages. Biochem Biophys Res Commun 2002; 293:274-83. [PMID: 12054596 PMCID: PMC2775714 DOI: 10.1016/s0006-291x(02)00138-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Peroxisome proliferator activated receptor-gamma (PPAR-gamma) is abundantly expressed in atherosclerotic lesions and is implicated in atherogenesis. The existence of three splice variants, PPAR-gamma 1, PPAR-gamma 2, and PPAR-gamma 3 has been established. Using monocyte-derived macrophages from cynomolgus monkeys, we demonstrate here the identification of two new PPAR-gamma exons, exon C and exon D, which splice together with already established exons A1, A2, and B in the 5(') terminal region to generate four novel PPAR-gamma subtypes, PPAR-gamma 4, -gamma 5, -gamma 6, and -gamma 7. PPAR-gamma 4 and gamma 5 were detected only in macrophages whereas gamma 6 and gamma 7 were expressed both in macrophages and adipose tissues. None of these novel isoforms were detected in muscle, kidney, and spleen from monkeys. We found sequences identical to exons C and D in the human genome database. These and all PPAR-gamma exons known to date are encoded by a single gene, located from region 10498 K to 10384 K on human chromosome 3. We cloned and expressed PPAR-gamma 1, PPAR-gamma 4, and PPAR-gamma 5 proteins in yeast using the expression vector pPICZB. As expected, all recombinant proteins showed a molecular weight of approximately 50 kDa. We also investigated the effect of a high-fat diet on the level of macrophage PPAR-gamma expression in monkeys. RT-PCR showed a significant increase in total PPAR-gamma and ABCA1 mRNA levels in macrophages of fat-fed monkeys (n=7) compared to those maintained on a normal diet (n=2). However, none of the novel isoforms seemed to be induced by fat-feeding. We used tetracycline-responsive expression vectors to obtain moderate expression of PPAR-gamma 4 and -gamma 5 in CHO cells. In these cells, expression of PPAR-gamma 5 but not -gamma 4 repressed the expression of ABCA1. Neither isoform modulated the expression of lipoprotein lipase. Our results suggest that individual PPAR-gamma isoforms may be responsible for unique tissue-specific biological effects and that PPAR-gamma 4 and -gamma 5 may modulate macrophage function and atherogenesis.
Collapse
Affiliation(s)
- Jiming Zhou
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA 52242, USA
- Department of Biotechnology, Sichuan University, Chengdu, Sichuan 610064, China
| | - Katina M. Wilson
- Department of Internal Medicine, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Jheem D. Medh
- Department of Biotechnology, Sichuan University, Chengdu, Sichuan 610064, China
- Department of Chemistry, Sc3112B, California State University at Northridge, 18111 Nordhoff Street, Northridge, CA 91330-8262, USA
- Corresponding author. Fax: +1-818-677-4068. (J.D. Medh)
| |
Collapse
|
45
|
Asakawa M, Takano H, Nagai T, Uozumi H, Hasegawa H, Kubota N, Saito T, Masuda Y, Kadowaki T, Komuro I. Peroxisome proliferator-activated receptor gamma plays a critical role in inhibition of cardiac hypertrophy in vitro and in vivo. Circulation 2002; 105:1240-6. [PMID: 11889020 DOI: 10.1161/hc1002.105225] [Citation(s) in RCA: 188] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
BACKGROUND Peroxisome proliferator-activated receptors (PPARs) are transcription factors of the nuclear receptor superfamily. It has been reported that the thiazolidinediones, which are antidiabetic agents and high-affinity ligands for PPARgamma, regulate growth of vascular cells. In the present study, we examined the role of PPARgamma in angiotensin II (Ang II)-induced hypertrophy of neonatal rat cardiac myocytes and in pressure overload-induced cardiac hypertrophy of mice. METHODS AND RESULTS Treatment of cultured cardiac myocytes with PPARgamma ligands such as troglitazone, pioglitazone, and rosiglitazone inhibited Ang II-induced upregulation of skeletal alpha-actin and atrial natriuretic peptide genes and an increase in cell surface area. Treatment of mice with a PPARgamma ligand, pioglitazone, inhibited pressure overload-induced increases in the heart weight-to-body weight ratio, wall thickness, and myocyte diameter in wild-type mice and an increase in the heart weight-to-body weight ratio in heterozygous PPARgamma-deficient mice. In contrast, pressure overload-induced increases in the heart weight-to-body weight ratio and wall thickness were more prominent in heterozygous PPARgamma-deficient mice than in wild-type mice. CONCLUSIONS These results suggest that the PPARgamma-dependent pathway is critically involved in the inhibition of cardiac hypertrophy.
Collapse
Affiliation(s)
- Masayuki Asakawa
- Department of Cardiovascular Science and Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
BACKGROUND The molecular basis of lipodystrophy, a syndrome associated with HIV antiretroviral (ARV) therapy, remains unknown. OBJECTIVES To examine whether ARV therapy might inhibit the expression of CD36, which is known to play an important role in fatty acid and glucose metabolism, and if this might contribute to the metabolic alterations associated with lipodystrophy. DESIGN The effects of ARV therapy on CD36 levels was examined in vivo in a prospective cohort of individuals treated with ARV therapy and in vitro in assays of human cell lines exposed to ARV drugs. METHODS Monocyte CD36 levels were assessed by flow cytometry at baseline and after 7 days of therapy in five healthy volunteers and 10 treatment-naive HIV-1-infected individuals. ARV therapy included protease inhibitors (ritonavir, nelfinavir or lopinavir/ritonavir). In addition, human cell lines (THP-1 and C32) were assessed for CD36 levels pre and post-ritonavir treatment. RESULTS Three of four healthy controls (one withdrew because of adverse effects) and 6 of 10 HIV-1-infected individuals had a 50 to > 90% decrease in monocyte CD36 levels after 7 days of therapy. One of ten HIV-infected subjects had a 30% decrease, and the remaining individuals had no change or an increase in CD36 levels. CD36 levels decreased significantly in human cell lines treated with ritonavir but not in those treated with zidovudine. CONCLUSIONS ARV therapy resulted in a marked decrease in CD36 in approximately 70% of our participants. Sustained ARV therapy-induced CD36 deficiency may contribute to insulin resistance and other metabolic complications of lipodystrophy.
Collapse
Affiliation(s)
- Lena Serghides
- Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
47
|
Molavi B, Rasouli N, Mehta JL. Peroxisome proliferator-activated receptor ligands as antiatherogenic agents: panacea or another Pandora's box? J Cardiovasc Pharmacol Ther 2002; 7:1-8. [PMID: 12000972 DOI: 10.1177/107424840200700i101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Peroxisome proliferator activated receptors (PPARs) are members of the nuclear receptor super family that modulate gene expression upon ligand activation. They are 3 major subtypes of PPARs: alpha, delta (also called beta), and gamma. PPAR-gamma is widely expressed in the cardiovascular system and is involved in the regulation of tissue inflammation and smooth muscle cell growth pathways as well as in lipoprotein metabolism and coagulation cascades. PPAR-gamma ligands of (e.g., rosigitazone and pioglitazone) have been shown to exert antiatherogenic effects both in vitro and in vivo. PPAR-alpha ligands (e.g., clofibrate and benzofibrate) modulate lipoprotein metabolism, and affect inflammation and coagulation cascade. These effects may be helpful in resolving the dilemma arising from studies that showed significant mortality and morbidity benefits of fibrates in the face of minimal changes in HDL-cholesterol levels. The role of PPAR-delta in atherogenesis remains largely unknown, although it appears that PPAR-delta activation affects lipoprotein metabolism. PPAR ligands appear to be promising agents in limiting atherosclerosis; however, large-scale clinical trials are required to assess their safety and efficacy before they can be added to the clinicians' arsenal of antiatherosclerotic agents.
Collapse
|
48
|
Ziouzenkova O, Perrey S, Marx N, Bacqueville D, Plutzky J. Peroxisome proliferator-activated receptors. Curr Atheroscler Rep 2002; 4:59-64. [PMID: 11772424 DOI: 10.1007/s11883-002-0063-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
An exciting and rapidly evolving area in vascular biology and atherosclerosis research over the past 3 years has been the establishment of peroxisome proliferator-activated receptor (PPAR) expression in the vascular and inflammatory cells, and the emerging picture of the roles these ligand-activated nuclear receptor/transcription factors might play in vascular biology and atherosclerosis. Such work is all the more compelling given the ongoing clinical use of PPAR activators in patients. Thiazolidinediones (PPAR-g agonists) are used as insulin sensitizers in diabetic patients known to be at extraordinarily high risk for cardiovascular disease, whereas fibrates (PPAR-a agonists) are used to treat dyslipidemia, particularly in the case of high triglycerides and low high-density lipoprotein cholesterol.
Collapse
Affiliation(s)
- Ouliana Ziouzenkova
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, 221 Longwood Avenue, LMRC 307, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
49
|
Benayoun L, Letuve S, Druilhe A, Boczkowski J, Dombret MC, Mechighel P, Megret J, Leseche G, Aubier M, Pretolani M. Regulation of peroxisome proliferator-activated receptor gamma expression in human asthmatic airways: relationship with proliferation, apoptosis, and airway remodeling. Am J Respir Crit Care Med 2001; 164:1487-94. [PMID: 11704601 DOI: 10.1164/ajrccm.164.8.2101070] [Citation(s) in RCA: 152] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Airway inflammation and alterations in cellular turnover are histopathologic features of asthma. We show that the expression of peroxisome proliferator-activated receptor gamma (PPAR gamma), a nuclear hormone receptor involved in cell activation, differentiation, proliferation, and/or apoptosis, is augmented in the bronchial submucosa, the airway epithelium, and the smooth muscle of steroid-untreated asthmatics, as compared with control subjects. This is associated with enhanced proliferation and apoptosis of airway epithelial and submucosal cells, as assessed by the immunodetection of the nuclear antigen Ki67, and of the cleaved form of caspase-3, respectively, and with signs of airway remodeling, including thickness of the subepithelial membrane (SBM) and collagen deposition. PPAR gamma expression in the epithelium correlates positively with SBM thickening and collagen deposition, whereas PPAR gamma expressing cells in the submucosa relate both to SBM thickening and to the number of proliferating cells. The intensity of PPAR gamma expression in the bronchial submucosa, the airway epithelium, and the smooth muscle is negatively related to FEV(1) values. Inhaled steroids alone, or associated with oral steroids, downregulate PPAR gamma expression in all the compartments, cell proliferation, SBM thickness, and collagen deposition, whereas they increase apoptotic cell numbers in the epithelium and the submucosa. Our findings have demonstrated that PPAR gamma (1) is a new indicator of airway inflammation and remodeling in asthma; (2) may be involved in extracellular matrix remodeling and submucosal cell proliferation; (3) is a target for steroid therapy.
Collapse
Affiliation(s)
- L Benayoun
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 408, Faculté de Médecine Xavier Bichat, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rosen ED, Spiegelman BM. PPARgamma : a nuclear regulator of metabolism, differentiation, and cell growth. J Biol Chem 2001; 276:37731-4. [PMID: 11459852 DOI: 10.1074/jbc.r100034200] [Citation(s) in RCA: 881] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- E D Rosen
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | |
Collapse
|