1
|
Zhang F, Fu Y, Zheng J, Lang L, Liang S, Wang J, Cai L, Zhang Y, Wang L, Zhu C, Wu R, Shu G, Jiang Q, Wang S. Conjugated Linoleic Acid (CLA) Mitigates High-Fat Diet (HFD)-Induced Mammary Gland Development Impairment of Pubertal Mice via Regulating CD36 Palmitoylation and Downstream JNK-ERK Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:1219-1230. [PMID: 39752534 DOI: 10.1021/acs.jafc.4c07453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Conjugated linoleic acid (CLA) is known for antiobesity. However, the role of CLA in regulating high-fat diet (HFD)-impaired pubertal mammary gland development remains undefined. Here, pubertal female mice and HC11 cells were treated with HFD or palmitic acid (PA), supplemented with or without CLA, respectively. We found that CLA prevented impaired mammary gland development in pubertal mice exposed to HFD. In vitro, c9, t11-CLA, but not t10, c12-CLA, promoted PA-suppressed HC11 proliferation, accompanied by hindered CD36 palmitoylation and localization on the plasma membrane. Moreover, c9, t11-CLA reduced the formation of the CD36/Fyn/Lyn complex and inhibited the JNK pathway while activated the ERK pathway in PA-treated HC11. In mechanism, the activation of the JNK pathway and the inhibition of ERK abolished the c9, t11-CLA-stimulated proliferation of PA-treated HC11. In vivo verification, CLA reduced the total and cell membrane CD36 palmitoylation, suppressed the formation of the CD36/FYN/LYN complex, and inhibited the JNK pathway but activated the ERK pathway in the mammary gland of HFD-fed mice. In conclusion, CLA mitigated HFD-impaired mammary gland development of pubertal mice and PA-suppressed HC11 proliferation via CD36 palmitoylation and the downstream JNK-ERK pathway. These data suggested the potential application of CLA in ameliorating obesity-impaired pubertal mammary gland development.
Collapse
Affiliation(s)
- Fenglin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Yiming Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Jisong Zheng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Limin Lang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Shuyi Liang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Junfeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Lilin Cai
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Yue Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Ruifan Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry and State Key Laboratory of Swine and Poultry Breeding Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China
- Yunfu Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Wens Foodstuff Group Co., Ltd., Yunfu 527400, P. R. China
| |
Collapse
|
2
|
Guo Y, Wei Z, Zhang Y, Cao J. The Effects of trans-10, cis-12 Conjugated Linoleic Acid on the Production Performance of Dairy Cows and the Expression and Transcription Regulation of Lipid Metabolism-Related Genes in Bovine Mammary Epithelial Cells. ACS OMEGA 2024; 9:34161-34174. [PMID: 39130591 PMCID: PMC11308042 DOI: 10.1021/acsomega.4c05532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 08/13/2024]
Abstract
Dietary fatty acids (FAs) determine the quality of dairy products. The trans-10, cis-12 conjugated linoleic acid (t10c12-CLA) is commonly considered an FA factor leading to milk fat depression syndrome (MFDs) in dairy cow. However, its effect on dairy cow performance and involvement in milk fat metabolism have been insufficiently explored. This study administered 136.17 g/day of rumen-protected CLA (RP-CLA) to dairy cows and found a diminution in milk fat percentage and a trend of increasing milk protein percentage on day 21 postpartum. Lactose content, milk yield, and net energy for lactation were unaffected. In the cell experiments, Oil Red O staining showed a notable increase in lipid droplets. Gene and protein expression analysis showed that 300 μM t10c12-CLA upregulated the expression of CD36, DGAT2, and ADRP, while downregulating the expression of ACACA, FASN, SREBP1, FABP3, FATP3, ACSL4, LPIN1, DGAT1, BTN1A1, XDH, SNAP23, and VAMP4. This provides a possible mechanistic pathway for the contradictory phenomenon of t10c12-CLA reducing milk fat while increasing lipid droplets. Overall, t10c12-CLA, as a long-chain fatty acid, can promote lipid droplet synthesis but may reduce milk fat by inhibiting lipid droplet fusion and secretion, FAs de novo synthesis, and triglyceride biosynthesis.
Collapse
Affiliation(s)
- Yuanyin Guo
- College
of Veterinary Medicine, China Agricultural
University, Beijing 100193, China
| | - Ziang Wei
- College
of Veterinary Medicine, China Agricultural
University, Beijing 100193, China
| | - Yi Zhang
- College
of Animal Science and Technology, China
Agricultural University, Beijing 100193, China
| | - Jie Cao
- College
of Veterinary Medicine, China Agricultural
University, Beijing 100193, China
| |
Collapse
|
3
|
Zhang F, Fu Y, Wang J, Lang L, Liang S, Zhang S, Wang L, Gao P, Shu G, Zhu C, Wu R, Jiang Q, Wang S. Conjugated linoleic acid (CLA) reduces intestinal fatty acid uptake and chylomicron formation in HFD-fed mice associated with the inhibition of DHHC7-mediated CD36 palmitoylation and the downstream ERK pathway. Food Funct 2024; 15:5000-5011. [PMID: 38618651 DOI: 10.1039/d4fo00099d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The anti-obesity effect of conjugated linoleic acid (CLA) has been well elucidated, but whether CLA affects fat deposition by regulating intestinal dietary fat absorption remains largely unknown. Thus, this study aimed to investigate the effects of CLA on intestinal fatty acid uptake and chylomicron formation and explore the possible underlying mechanisms. We found that CLA supplementation reduced the intestinal fat absorption in HFD (high fat diet)-fed mice accompanied by the decreased serum TG level, increased fecal lipids and decreased intestinal expression of ApoB48 and MTTP. Correspondingly, c9, t11-CLA, but not t10, c12-CLA induced the reduction of fatty acid uptake and TG content in PA (palmitic acid)-treated MODE-K cells. In the mechanism of fatty acid uptake, c9, t11-CLA inhibited the binding of CD36 with palmitoyltransferase DHHC7, thus leading to the decreases of CD36 palmitoylation level and localization on the cell membrane of the PA-treated MODE-K cells. In the mechanism of chylomicron formation, c9, t11-CLA inhibited the formation of the CD36/FYN/LYN complex and the activation of the ERK pathway in the PA-treated MODE-K cells. In in vivo verification, CLA supplementation reduced the DHHC7-mediated total and cell membrane CD36 palmitoylation and suppressed the formation of the CD36/FYN/LYN complex and the activation of the ERK pathway in the jejunum of HFD-fed mice. Altogether, these data showed that CLA reduced intestinal fatty acid uptake and chylomicron formation in HFD-fed mice associated with the inhibition of DHHC7-mediated CD36 palmitoylation and the downstream ERK pathway.
Collapse
Affiliation(s)
- Fenglin Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Yiming Fu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Junfeng Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Limin Lang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Shuyi Liang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Shilei Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Ruifan Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control and National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, P. R. China.
- Yunfu Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Wens Foodstuff Group Co., Ltd.,Yunfu 527400, P. R. China
| |
Collapse
|
4
|
Lin W, Song H, Shen J, Wang J, Yang Y, Yang Y, Cao J, Xue L, Zhao F, Xiao T, Lin R. Functional role of skeletal muscle-derived interleukin-6 and its effects on lipid metabolism. Front Physiol 2023; 14:1110926. [PMID: 37555019 PMCID: PMC10405179 DOI: 10.3389/fphys.2023.1110926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 07/06/2023] [Indexed: 08/10/2023] Open
Abstract
The detrimental impact of obesity on human health is increasingly evident with the rise in obesity-related diseases. Skeletal muscle, the crucial organ responsible for energy balance metabolism, plays a significant role as a secretory organ by releasing various myokines. Among these myokines, interleukin 6 (IL-6) is closely associated with skeletal muscle contraction. IL-6 triggers the process of lipolysis by mobilizing energy-storing adipose tissue, thereby providing energy for physical exercise. This phenomenon also elucidates the health benefits of regular exercise. However, skeletal muscle and adipose tissue maintain a constant interaction, both directly and indirectly. Direct interaction occurs through the accumulation of excess fat within skeletal muscle, known as ectopic fat deposition. Indirect interaction takes place when adipose tissue is mobilized to supply the energy for skeletal muscle during exercise. Consequently, maintaining a functional balance between skeletal muscle and adipose tissue becomes paramount in regulating energy metabolism and promoting overall health. IL-6, as a representative cytokine, participates in various inflammatory responses, including non-classical inflammatory responses such as adipogenesis. Skeletal muscle influences adipogenesis through paracrine mechanisms, primarily by secreting IL-6. In this research paper, we aim to review the role of skeletal muscle-derived IL-6 in lipid metabolism and other physiological activities, such as insulin resistance and glucose tolerance. By doing so, we provide valuable insights into the regulatory function of skeletal muscle-derived myokines in lipid metabolism.
Collapse
Affiliation(s)
- Weimin Lin
- *Correspondence: Weimin Lin, ; Ruiyi Lin,
| | | | | | | | | | | | | | | | | | | | - Ruiyi Lin
- College of Animal Sciences (College of Bee Science), Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
5
|
Multiple biological activities and biosynthesis mechanisms of specific conjugated linoleic acid isomers and analytical methods for prospective application. Food Chem 2023; 409:135257. [PMID: 36584529 DOI: 10.1016/j.foodchem.2022.135257] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 11/27/2022] [Accepted: 12/18/2022] [Indexed: 12/30/2022]
Abstract
Conjugated linoleic acid (CLA) is a collective term for the octadecadienoic acid isomers containing conjugated double bonds. This article reviewed CLA isomers from biological activities, biosynthesis mechanisms and analytical methods. The biological activities of CLA isomers in anti-obesity, cardiovascular protection, diabetes management and anti-cancer in vitro and in vivo were mainly reviewed. More attention has been paid to the production of the specific CLA isomer due to its biological activity. The biosynthesis methods of CLA isomers, such as dietary modification in ruminants and fermentation by microorganisms & enzymes, were systematically introduced. A rapid, accurate and economic analysis method will promote the research in both biological activities and biosynthesis mechanisms of CLA isomers. The merits of UV spectrometry, GC, HPLC, MS and CE used in the analysis of CLA isomers were also compared in detail. This paper aims to put into perspective the current status and future trends on CLA isomers.
Collapse
|
6
|
Wan Q, Huang B, Li T, Xiao Y, He Y, Du W, Wang BZ, Dakin GF, Rosenbaum M, Goncalves MD, Chen S, Leong KW, Qiang L. Selective targeting of visceral adiposity by polycation nanomedicine. NATURE NANOTECHNOLOGY 2022; 17:1311-1321. [PMID: 36456644 DOI: 10.1038/s41565-022-01249-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/03/2022] [Indexed: 06/17/2023]
Abstract
Obesity is a pandemic health problem with poor solutions, especially for targeted treatment. Here we develop a polycation-based nanomedicine polyamidoamine generation 3 (P-G3) that-when delivered intraperitoneally-selectively targets visceral fat due to its high charge density. Moreover, P-G3 treatment of obese mice inhibits visceral adiposity, increases energy expenditure, prevents obesity and alleviates the associated metabolic dysfunctions. In vitro adipogenesis models and single-cell RNA sequencing revealed that P-G3 uncouples adipocyte lipid synthesis and storage from adipocyte development to create adipocytes that possess normal functions but are deficient in hypertrophic growth, at least through synergistically modulating nutrient-sensing signalling pathways. The visceral fat distribution of P-G3 is enhanced by modifying P-G3 with cholesterol to form lipophilic nanoparticles, which is effective in treating obesity. Our study highlights a strategy to target visceral adiposity and suggests that cationic nanomaterials could be exploited for treating metabolic diseases.
Collapse
Affiliation(s)
- Qianfen Wan
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Baoding Huang
- Department of Orthopaedic Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Ying He
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Wen Du
- Department of Medicine, Columbia University, New York, NY, USA
| | - Branden Z Wang
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Gregory F Dakin
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Michael Rosenbaum
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Pediatrics, Columbia University, New York, NY, USA
| | | | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Li Qiang
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
7
|
Osborn LJ, Schultz K, Massey W, DeLucia B, Choucair I, Varadharajan V, Banerjee R, Fung K, Horak AJ, Orabi D, Nemet I, Nagy LE, Wang Z, Allende DS, Willard BB, Sangwan N, Hajjar AM, McDonald C, Ahern PP, Hazen SL, Brown JM, Claesen J. A gut microbial metabolite of dietary polyphenols reverses obesity-driven hepatic steatosis. Proc Natl Acad Sci U S A 2022; 119:e2202934119. [PMID: 36417437 PMCID: PMC9860326 DOI: 10.1073/pnas.2202934119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/13/2022] [Indexed: 11/24/2022] Open
Abstract
The molecular mechanisms by which dietary fruits and vegetables confer cardiometabolic benefits remain poorly understood. Historically, these beneficial properties have been attributed to the antioxidant activity of flavonoids. Here, we reveal that the host metabolic benefits associated with flavonoid consumption hinge, in part, on gut microbial metabolism. Specifically, we show that a single gut microbial flavonoid catabolite, 4-hydroxyphenylacetic acid (4-HPAA), is sufficient to reduce diet-induced cardiometabolic disease (CMD) burden in mice. The addition of flavonoids to a high fat diet heightened the levels of 4-HPAA within the portal plasma and attenuated obesity, and continuous delivery of 4-HPAA was sufficient to reverse hepatic steatosis. The antisteatotic effect was shown to be associated with the activation of AMP-activated protein kinase α (AMPKα). In a large survey of healthy human gut metagenomes, just over one percent contained homologs of all four characterized bacterial genes required to catabolize flavonols into 4-HPAA. Our results demonstrate the gut microbial contribution to the metabolic benefits associated with flavonoid consumption and underscore the rarity of this process in human gut microbial communities.
Collapse
Affiliation(s)
- Lucas J. Osborn
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
| | - Karlee Schultz
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- College of Arts and Sciences, John Carroll University, University Heights, OH44118
| | - William Massey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
| | - Beckey DeLucia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Ibrahim Choucair
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Venkateshwari Varadharajan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Rakhee Banerjee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Kevin Fung
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Anthony J. Horak
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Danny Orabi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
- Department of General Surgery, Cleveland Clinic, Cleveland, OH44195
| | - Ina Nemet
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Laura E. Nagy
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
- Department of Inflammation and Immunity, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Daniela S. Allende
- Robert J. Tomsich Pathology and Laboratory Medicine Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Belinda B. Willard
- Mass Spectrometry Core, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Naseer Sangwan
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Adeline M. Hajjar
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Christine McDonald
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
- Department of Inflammation and Immunity, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
| | - Philip P. Ahern
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Department of Cardiovascular Medicine, Heart Vascular, and Thoracic Institute Cleveland Clinic, Cleveland, OH44195
| | - J. Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
| | - Jan Claesen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Center for Microbiome and Human Health, Lerner Research Institute of the Cleveland Clinic, Cleveland, OH44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH44195
| |
Collapse
|
8
|
Manaserh IH, Bledzka KM, Junker A, Grondolsky J, Schumacher SM. A Cardiac Amino-Terminal GRK2 Peptide Inhibits Maladaptive Adipocyte Hypertrophy and Insulin Resistance During Diet-Induced Obesity. JACC Basic Transl Sci 2022; 7:563-579. [PMID: 35818501 PMCID: PMC9270572 DOI: 10.1016/j.jacbts.2022.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 12/04/2022]
Abstract
Heart disease remains the leading cause of death, and mortality rates positively correlate with the presence of obesity and diabetes. Despite the correlation between cardiac and metabolic dysregulation, the mechanistic pathway(s) of interorgan crosstalk still remain undefined. This study reveals that cardiac-restricted expression of an amino-terminal peptide of GRK2 (βARKnt) preserves systemic and cardiac insulin responsiveness, and protects against adipocyte maladaptive hypertrophy in a diet-induced obesity model. These data suggest a cardiac-driven mechanism to ameliorate maladaptive cardiac remodeling and improve systemic metabolic homeostasis that may lead to new treatment modalities for cardioprotection in obesity and obesity-related metabolic syndromes.
Collapse
Key Words
- AS160, Akt substrate of 160 kilodaltons
- BAT, brown adipose tissue
- GRK2
- GRK2, G protein-coupled receptor kinase 2
- HFD, high-fat diet
- HOMA-IR, homeostatic model assessment of insulin resistance
- NLC, nontransgenic littermate control
- NP, natriuretic peptide
- NPR, natriuretic peptide receptor
- RER, respiratory exchange ratio
- T2D, type II diabetes
- Tg, transgenic
- beiging
- cardioprotection
- gWAT, gonadal white adipose tissue
- mTOR, mechanistic target of rapamycin protein kinase
- metabolism
- obesity
- βARKct, cardiac restricted expression of C-terminus domain of GRK2
- βARKnt, cardiac-restricted expression of N-terminus domain of GRK2
Collapse
Affiliation(s)
- Iyad H. Manaserh
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Kamila M. Bledzka
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Alex Junker
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jessica Grondolsky
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sarah M. Schumacher
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
9
|
Massey W, Osborn LJ, Banerjee R, Horak A, Fung KK, Orabi D, Chan ER, Sangwan N, Wang Z, Brown JM. Flavin-Containing Monooxygenase 3 (FMO3) Is Critical for Dioxin-Induced Reorganization of the Gut Microbiome and Host Insulin Sensitivity. Metabolites 2022; 12:364. [PMID: 35448550 PMCID: PMC9029240 DOI: 10.3390/metabo12040364] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/18/2022] [Accepted: 04/07/2022] [Indexed: 02/07/2023] Open
Abstract
Exposure to some environmental pollutants can have potent endocrine-disrupting effects, thereby promoting hormone imbalance and cardiometabolic diseases such as non-alcoholic fatty liver disease (NAFLD), diabetes, and cardiorenal diseases. Recent evidence also suggests that many environmental pollutants can reorganize the gut microbiome to potentially impact these diverse human diseases. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is among the most potent endocrine-disrupting dioxin pollutants, yet our understanding of how TCDD impacts the gut microbiome and systemic metabolism is incompletely understood. Here, we show that TCDD exposure in mice profoundly stimulates the hepatic expression of flavin-containing monooxygenase 3 (Fmo3), which is a hepatic xenobiotic metabolizing enzyme that is also responsible for the production of the gut microbiome-associated metabolite trimethylamine N-oxide (TMAO). Interestingly, an enzymatic product of FMO3 (TMAO) has been associated with the same cardiometabolic diseases that these environmental pollutants promote. Therefore, here, we examined TCDD-induced alterations in the gut microbiome, host liver transcriptome, and glucose tolerance in Fmo3+/+ and Fmo3-/- mice. Our results show that Fmo3 is a critical component of the transcriptional response to TCDD, impacting the gut microbiome, host liver transcriptome, and systemic glucose tolerance. Collectively, this work uncovers a previously underappreciated role for Fmo3 in integrating diet-pollutant-microbe-host interactions.
Collapse
Affiliation(s)
- William Massey
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lucas J. Osborn
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Rakhee Banerjee
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Anthony Horak
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Kevin K. Fung
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
| | - Danny Orabi
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of General Surgery, Cleveland Clinic, Cleveland, OH 44195, USA
| | - E. Ricky Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Naseer Sangwan
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Microbial Sequencing & Analytics Core Facility, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - J. Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA; (W.M.); (L.J.O.); (R.B.); (A.H.); (K.K.F.); (D.O.); (Z.W.)
- Center for Microbiome & Human Health, Cleveland Clinic, Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
10
|
Chirivi M, Rendon CJ, Myers MN, Prom CM, Roy S, Sen A, Lock AL, Contreras GA. Lipopolysaccharide induces lipolysis and insulin resistance in adipose tissue from dairy cows. J Dairy Sci 2021; 105:842-855. [PMID: 34696909 DOI: 10.3168/jds.2021-20855] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/05/2021] [Indexed: 01/05/2023]
Abstract
Intense and protracted adipose tissue (AT) fat mobilization increases the risk of metabolic and inflammatory periparturient diseases in dairy cows. This vulnerability increases when cows have endotoxemia-common during periparturient diseases such as mastitis, metritis, and pneumonia-but the mechanisms are unknown. Fat mobilization intensity is determined by the balance between lipolysis and lipogenesis. Around parturition, the rate of lipolysis surpasses that of lipogenesis, leading to enhanced free fatty acid release into the circulation. We hypothesized that exposure to endotoxin (ET) increases AT lipolysis by activation of classic and inflammatory lipolytic pathways and reduction of insulin sensitivity. In experiment 1, subcutaneous AT (SCAT) explants were collected from periparturient (n = 12) Holstein cows at 11 ± 3.6 d (mean ± SE) before calving, and 6 ± 1 d and 13 ± 1.4 d after parturition. Explants were treated with the endotoxin lipopolysaccharide (LPS; 20 µg/mL; basal = 0 µg/mL) for 3 h. The effect of LPS on lipolysis was assessed in the presence of the β-adrenergic agonist and promoter of lipolysis isoproterenol (ISO; 1 µM; LPS+ISO). In experiment 2, SCAT explants were harvested from 24 nonlactating, nongestating multiparous Holstein dairy cows and exposed to the same treatments as in experiment 1 for 3 and 7 h. The effect of LPS on the antilipolytic responses induced by insulin (INS = 1 µL/L, LPS+INS) was established during ISO stimulation [ISO+INS, LPS+ISO+INS]. The characterization of lipolysis included the quantification of glycerol release and the assessment of markers of lipase activity [adipose triglyceride lipase (ATGL), hormone-sensitive lipase (HSL), and phosphorylated HSL Ser563 (pHSL)], and insulin pathway activation (AKT, pAKT) using capillary electrophoresis. Inflammatory gene networks were evaluated by real-time quantitative PCR. In periparturient cows, LPS increased AT lipolysis by 67 ± 12% at 3 h across all time points compared with basal. In nonlactating cows, LPS was an effective lipolytic agent at 3 h and 7 h, increasing glycerol release by 115 ± 18% and 68.7 ± 16%, respectively, relative to basal. In experiment 2, LPS enhanced ATGL activity with minimal HSL activation at 3 h. In contrast, at 7 h, LPS increased HSL phosphorylation (i.e., HSL activity) by 123 ± 11%. The LPS-induced HSL lipolytic activity at 7 h coincided with the activation of the MEK/ERK inflammatory pathway. In experiment 2, INS reduced the lipolytic effect of ISO (ISO+INS: -63 ± 18%) and LPS (LPS+INS: -45.2 ± 18%) at 3 h. However, the antilipolytic effect of INS was lost in the presence of LPS at 7 h (LPS+INS: -16.3 ± 16%) and LPS+ISO+INS at 3 and 7 h (-3.84 ± 23.6% and -21.2 ± 14.6%). Accordingly, LPS reduced pAKT:AKT (0.11 ± 0.07) compared with basal (0.18 ± 0.05) at 7 h. Our results indicated that exposure to LPS activated the classic and inflammatory lipolytic pathways and reduced insulin sensitivity in SCAT. These data provide evidence that during endotoxemia, dairy cows may be more susceptible to lipolysis dysregulation and loss of adipocyte sensitivity to the antilipolytic action of insulin.
Collapse
Affiliation(s)
- Miguel Chirivi
- Department of Large Animal Clinical Science, Michigan State University, East Lansing 48824
| | - C Javier Rendon
- Department of Large Animal Clinical Science, Michigan State University, East Lansing 48824
| | - Madison N Myers
- Department of Large Animal Clinical Science, Michigan State University, East Lansing 48824
| | - Crystal M Prom
- Department of Animal Sciences, Michigan State University, East Lansing 48824
| | - Sambit Roy
- Department of Animal Sciences, Michigan State University, East Lansing 48824
| | - Aritro Sen
- Department of Animal Sciences, Michigan State University, East Lansing 48824
| | - Adam L Lock
- Department of Animal Sciences, Michigan State University, East Lansing 48824
| | - G Andres Contreras
- Department of Large Animal Clinical Science, Michigan State University, East Lansing 48824.
| |
Collapse
|
11
|
Dermal white adipose tissue: Much more than a metabolic, lipid-storage organ? Tissue Cell 2021; 71:101583. [PMID: 34171520 DOI: 10.1016/j.tice.2021.101583] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/25/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022]
Abstract
The role of dermal white adipose tissue (dWAT) has emerged in the biomedical science as an ancillary fat district in the derma without a defined and distinct function respect to the subcutaneous adipose tissue (sWAT). Despite some evidence describing dWAT as an immune-competent compartment, particularly engaged in wound repair, very few reports dealing with dWAT has elucidated its major modulatory role within the skin biology. Whereas an increasing bulk of evidence allows researcher to describe the main activity of sWAT, in humans dWAT is not properly a separated fat compartment and therefore scarcely considered in the scientific debate. Due to its strategic position between epidermis and sWAT, dermal fat might play a much more intriguing role than expected. This review tries to shed light on this issue, by expanding the debate about a possible role of dWAT in skin physiology.
Collapse
|
12
|
Supplementing conjugated linoleic acid (CLA) in breeder hens diet increased CLA incorporation in liver and alters hepatic lipid metabolism in chick offspring. Br J Nutr 2021; 127:1443-1454. [PMID: 33658091 DOI: 10.1017/s0007114521000763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
This experiment was designed to investigate the effect of supplementing conjugated linoleic acid (CLA) in breeder hens diet on development and hepatic lipid metabolism of chick offspring. Hy-Line Brown breeder hens were allocated into two groups, supplemented with 0 (CT) or 0.5% CLA for 8 weeks. Offspring chicks were grouped according to the mother generation and fed for 7 days. CLA treatment had no significant influence on development, egg quality, and fertility of breeder hens, but darkened the egg yolks in shade and increased yolk sac mass compared to CT group. Addition of CLA resulted in increased body mass and liver mass, and decreased deposition of subcutaneous adipose tissue in chick offspring. The serum triglyceride (TG) and cholesterol (TC) levels of chick offspring were decreased in CLA group. CLA treatment increased the incorporation of both CLA isomers (c9t11 and t10c12) in liver of chick offspring, accompanied by the decreased hepatic TG levels, related to the significant reduction of fatty acid synthase (FAS) and acetyl-CoA carboxylase (ACC) enzyme activities and the increased of carnitine palmitoyltransferase-1 (CPT1) enzyme activity. Meanwhile, CLA treatment reduced the mRNA expression of genes related to fatty acid biosynthesis (FAS, ACC, and sterol regulatory element-binding protein-1c), and induced the expression of genes related to β-oxidative (CPT1, AMP-activated protein kinase, and peroxisome proliferator-activated receptor α) in chick offspring liver. In summary, the addition of CLA in breeder hens diet significantly increased incorporation of CLA in liver of chick offspring, which further regulate hepatic lipid metabolism.
Collapse
|
13
|
Neumann CKA, Silver DJ, Venkateshwari V, Zhang R, Traughber CA, Przybycin C, Bayik D, Smith JD, Lathia JD, Rini BI, Brown JM. MBOAT7-driven phosphatidylinositol remodeling promotes the progression of clear cell renal carcinoma. Mol Metab 2020; 34:136-145. [PMID: 32180553 PMCID: PMC7033598 DOI: 10.1016/j.molmet.2020.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 01/21/2020] [Indexed: 12/28/2022] Open
Abstract
Objective The most common kidney cancer, clear cell renal cell carcinoma (ccRCC), is closely associated with obesity. The “clear cell” variant of RCC gets its name from the large lipid droplets that accumulate in the tumor cells. Although renal lipid metabolism is altered in ccRCC, the mechanisms and lipids driving this are not well understood. Methods We used shotgun lipidomics in human ccRCC tumors and matched normal adjacent renal tissue. To assess MBOAT7s gene expression across tumor severity, we examined histologically graded human ccRCC samples. We then utilized genome editing in ccRCC cell lines to understand the role of MBOAT7 in ccRCC progression. Results We identified a lipid signature for ccRCC that includes an increase in arachidonic acid-enriched phosphatidylinositols (AA-PI). In parallel, we found that ccRCC tumors have increased expression of acyltransferase enzyme membrane bound O-acyltransferase domain containing 7 (MBOAT7) that contributes to AA-PI synthesis. In ccRCC patients, MBOAT7 expression increases with tumor grade, and increased MBOAT7 expression correlates with poor survival. Genetic deletion of MBOAT7 in ccRCC cells decreases proliferation and induces cell cycle arrest, and MBOAT7−/− cells fail to form tumors in vivo. RNAseq of MBOAT7−/− cells identified alterations in cell migration and extracellular matrix organization that were functionally validated in migration assays. Conclusions This study highlights the accumulation of AA-PI in ccRCC and demonstrates a novel way to decrease the AA-PI pool in ccRCC by limiting MBOAT7. Our data reveal that metastatic ccRCC is associated with altered AA-PI metabolism and identify MBOAT7 as a novel target in advanced ccRCC.
Collapse
Affiliation(s)
- Chase K A Neumann
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Daniel J Silver
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Varadharajan Venkateshwari
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Renliang Zhang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - C Alicia Traughber
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Christopher Przybycin
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Defne Bayik
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jonathan D Smith
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Justin D Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA; Rose Ella Burkhardt Brain Tumor and Neuro-Oncology Center, Cleveland Clinic, Cleveland, OH, 44195, USA; Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Brian I Rini
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Cleveland, OH, 44195, USA; Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - J Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA; Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA; Center for Microbiome and Human Health, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA.
| |
Collapse
|
14
|
Chirumbolo S. Oxidative Stress, Nutrition and Cancer: Friends or Foes? World J Mens Health 2020; 39:19-30. [PMID: 32202081 PMCID: PMC7752511 DOI: 10.5534/wjmh.190167] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
The relationship between cancer and nutrition, as well as nutrition and oxidative stress, shares puzzling aspects that current research is investigating as the possible components of an intriguing regulating mechanism involving the complex interplay between adipose tissue and other compartments. Along the very recent biological evolution, humans underwent a rapid change in their lifestyles and henceforth the role of the adipocytes earned a much more complex task in the fine tuning of the tissue microenvironment. A lipidic signaling language probably evolved in association with the signaling role of reactive oxygen species, which gained a fundamental part in the regulation of cell stem and plasticity. The possible relationship with cancer onset might have some causative mechanism in the impairment of this complex task, usually deregulated by drastic changes in one's own lifestyle and dietary habit. This review tries to address this issue.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| |
Collapse
|
15
|
Shi W, Hegeman MA, Doncheva A, van der Stelt I, Bekkenkamp‐Grovenstein M, van Schothorst EM, Brenner C, de Boer VCJ, Keijer J. Transcriptional Response of White Adipose Tissue to Withdrawal of Vitamin B3. Mol Nutr Food Res 2019; 63:e1801100. [PMID: 30990964 PMCID: PMC6618275 DOI: 10.1002/mnfr.201801100] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/11/2019] [Indexed: 01/24/2023]
Abstract
SCOPE Distinct markers for mild vitamin B3 deficiency are lacking. To identify these, the molecular responses of white adipose tissue (WAT) to vitamin B3 withdrawal are examined. METHODS AND RESULTS A dietary intervention is performed in male C57BL/6JRccHsd mice, in which a diet without nicotinamide riboside (NR) is compared to a diet with NR at the recommended vitamin B3 level. Both diets contain low but adequate level of tryptophan. Metabolic flexibility and systemic glucose tolerance are analyzed and global transcriptomics, qRT-PCR, and histology of epididymal WAT (eWAT) are performed. A decreased insulin sensitivity and a shift from carbohydrate to fatty acid oxidation in response to vitamin B3 withdrawal are observed. This is consistent with molecular changes in eWAT, including an activated MEK/ERK signaling, a lowering of glucose utilization markers, and an increase in makers of fatty acid catabolism, possibly related to the consistent lower expression of mitochondrial electron transport complexes. The synthesis pathway of tetrahydropteridine (BH4), an essential cofactor for neurotransmitter synthesis, is transcriptionally activated. Genes marking these processes are technically validated. CONCLUSION The downregulation of Anp32a, Tnk2 and the upregulation of Mapk1, Map2k1, Qdpr, Mthfs, and Mthfsl are proposed as a WAT transcriptional signature marker for mild vitamin B3 deficiency.
Collapse
Affiliation(s)
- Wenbiao Shi
- Human and Animal PhysiologyWageningen UniversityPO Box 3386700AHWageningenThe Netherlands
| | - Maria A. Hegeman
- Human and Animal PhysiologyWageningen UniversityPO Box 3386700AHWageningenThe Netherlands
- Educational Consultancy & Professional DevelopmentFaculty of Social and Behavioural Sciences, Utrecht University3584CSUtrechtThe Netherlands
| | - Atanaska Doncheva
- Human and Animal PhysiologyWageningen UniversityPO Box 3386700AHWageningenThe Netherlands
| | - Inge van der Stelt
- Human and Animal PhysiologyWageningen UniversityPO Box 3386700AHWageningenThe Netherlands
| | | | | | - Charles Brenner
- Department of BiochemistryCarver College of Medicine, University of IowaIowa CityIA52242USA
| | - Vincent C. J. de Boer
- Human and Animal PhysiologyWageningen UniversityPO Box 3386700AHWageningenThe Netherlands
| | - Jaap Keijer
- Human and Animal PhysiologyWageningen UniversityPO Box 3386700AHWageningenThe Netherlands
| |
Collapse
|
16
|
Zhu W, Buffa JA, Wang Z, Warrier M, Schugar R, Shih DM, Gupta N, Gregory JC, Org E, Fu X, Li L, DiDonato JA, Lusis AJ, Brown JM, Hazen SL. Flavin monooxygenase 3, the host hepatic enzyme in the metaorganismal trimethylamine N-oxide-generating pathway, modulates platelet responsiveness and thrombosis risk. J Thromb Haemost 2018; 16:1857-1872. [PMID: 29981269 PMCID: PMC6156942 DOI: 10.1111/jth.14234] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 06/26/2018] [Indexed: 12/21/2022]
Abstract
Essentials Microbe-dependent production of trimethylamine N-oxide (TMAO) contributes to thrombosis risk. The impact of host flavin monooxygenase 3 (FMO3) modulation on platelet function is unknown. Genetic manipulation of FMO3 in mice alters systemic TMAO levels and thrombosis potential. Genetic manipulation of FMO3 is associated with alteration of gut microbial community structure. SUMMARY Background Gut microbes play a critical role in the production of trimethylamine N-oxide (TMAO), an atherogenic metabolite that impacts platelet responsiveness and thrombosis potential. Involving both microbe and host enzymatic machinery, TMAO generation utilizes a metaorganismal pathway, beginning with ingestion of trimethylamine (TMA)-containing dietary nutrients such as choline, phosphatidylcholine and carnitine, which are abundant in a Western diet. Gut microbial TMA lyases use these nutrients as substrates to produce TMA, which upon delivery to the liver via the portal circulation, is converted into TMAO by host hepatic flavin monooxygenases (FMOs). Gut microbial production of TMA is rate limiting in the metaorganismal TMAO pathway because hepatic FMO activity is typically in excess. Objectives FMO3 is the major FMO responsible for host generation of TMAO; however, a role for FMO3 in altering platelet responsiveness and thrombosis potential in vivo has not yet been explored. Methods The impact of FMO3 suppression (antisense oligonucleotide-targeting) and overexpression (as transgene) on plasma TMAO levels, platelet responsiveness and thrombosis potential was examined using a murine FeCl3 -induced carotid artery injury model. Cecal microbial composition was examined using 16S analyses. Results Modulation of FMO3 directly impacts systemic TMAO levels, platelet responsiveness and rate of thrombus formation in vivo. Microbial composition analyses reveal taxa whose proportions are associated with both plasma TMAO levels and in vivo thrombosis potential. Conclusions The present studies demonstrate that host hepatic FMO3, the terminal step in the metaorganismal TMAO pathway, participates in diet-dependent and gut microbiota-dependent changes in both platelet responsiveness and thrombosis potential in vivo.
Collapse
Affiliation(s)
- W. Zhu
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. A. Buffa
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Z. Wang
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - M. Warrier
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - R. Schugar
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - D. M. Shih
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - N. Gupta
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. C. Gregory
- Departments of Human Genetics and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - E. Org
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - X. Fu
- Departments of Human Genetics and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - L. Li
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. A. DiDonato
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - A. J. Lusis
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - J. M. Brown
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Departments of Human Genetics and Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - S. L. Hazen
- Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
- Heart and Vascular Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
17
|
Sadri H, Saremi B, Dänicke S, Rehage J, Mielenz M, Hosseini A, Sauerwein H. Lactation-related changes in tissue expression of PEDF in dairy cows. Domest Anim Endocrinol 2018; 64:93-101. [PMID: 29758402 DOI: 10.1016/j.domaniend.2018.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 12/21/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is evolving as metabolic regulatory protein. Albeit mostly considered in only pathological conditions related to excess energy intake resulting in obesity and insulin resistance, PEDF is likely to be involved in other physiological processes such as the homeorhetic adaptation of metabolism to lactation. We aimed to characterize the expression of PEDF and its association to the concomitant mobilization of body reserves during lactation in nonobese subjects. This mobilization is particularly distinct in dairy cows, and we therefore assessed the mRNA expression of PEDF and its putative receptors in different tissues in 2 trials with dairy cows fed with or without conjugated linoleic acids (CLAs). Conjugated linoleic acids depress milk fat synthesis and may thus reduce the drain of energy via milk. In pluriparous cows, the serum PEDF concentrations and the mRNA abundance in subcutaneous adipose tissue (scAT), as well as the hepatic and scAT mRNA abundance of the putative receptors, adipose triglyceride lipase, and laminin receptor 1, changed over time of sampling (day -21 until day 252 relative to calving). Conjugated linoleic acid treatment was associated with reduced PEDF concentrations in serum and lower PEDF mRNA abundance in scAT on day 21 postpartum. Comparing different tissues from primiparous cows, PEDF mRNA was highest in the liver, followed by scAT, visceral adipose tissue (AT), and mammary gland, and lowest in the muscle. Significant changes in PEDF expression with time of sampling were limited to AT in primiparous and pluriparous cows. Our data support a regulatory role for PEDF. The similarities between the time course of the serum concentrations of PEDF and its mRNA abundance in scAT may point to a regulatory role for AT rather than the liver for PEDF in dairy cows.
Collapse
Affiliation(s)
- H Sadri
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran; Institute of Animal Science, Physiology & Hygiene Unit, University of Bonn, Bonn 53111, Germany
| | - B Saremi
- Institute of Animal Science, Physiology & Hygiene Unit, University of Bonn, Bonn 53111, Germany
| | - S Dänicke
- Institute of Animal Nutrition, Friedrich-Loeffler-Institute (FLI), Federal Research Institute for Animal Health, Braunschweig 38116, Germany
| | - J Rehage
- Clinic for Cattle, University of Veterinary Medicine, Hannover 30173, Germany
| | - M Mielenz
- Institute of Nutritional Physiology "Oskar Kellner", Leibniz Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, Dummerstorf 18196, Germany
| | - A Hosseini
- Department of Clinical Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran
| | - H Sauerwein
- Institute of Animal Science, Physiology & Hygiene Unit, University of Bonn, Bonn 53111, Germany.
| |
Collapse
|
18
|
von Frankenberg AD, Reis AF, Gerchman F. Relationships between adiponectin levels, the metabolic syndrome, and type 2 diabetes: a literature review. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2017; 61:614-622. [PMID: 29412387 PMCID: PMC10522055 DOI: 10.1590/2359-3997000000316] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 08/09/2017] [Indexed: 11/22/2022]
Abstract
Elevated hepatic glucose production, impaired insulin secretion, and insulin resistance - abnormalities of glucose metabolism typically found in subjects with obesity - are major factors underlying the pathogenesis of type 2 diabetes (DM2) and the metabolic syndrome (MS). Adiponectin is a major regulator of glucose and lipid homeostasis via its insulin-sensitizing properties, and lower levels seems to be associated with the development of DM2 and MS. The purpose of this review is to clarify the mechanisms whereby adiponectin relates to the development of DM2 and MS and the association between polymorphisms of the adiponectin gene, circulating levels of the hormone, and its relationships with DM2. In addition, the impact of dietary lipids in the circulating levels of adiponectin will be addressed. According to the literature, circulating adiponectin levels seem to decrease as the number of MS components increases. Lower adiponectin concentrations are associated with higher intra-abdominal fat content. Therefore, adiponectin could link intra-abdominal fat with insulin resistance and development of MS. Therapeutic strategies that target the MS and its components, such as lifestyle modification through physical activity and weight loss, have been shown to increase adiponectin concentrations. Possible roles of diets containing either low or high amounts of fat, or different types of fat, have been analyzed in several studies, with heterogeneous results. Supplementation with n-3 PUFA modestly increases adiponectin levels, whereas conjugated linoleic acid supplementation appears to reduce concentrations when compared with unsaturated fatty acid supplementation used as an active placebo.
Collapse
Affiliation(s)
- Anize Delfino von Frankenberg
- Universidade Federal do Rio Grande do SulFaculdade de MedicinaPorto AlegreRSBrasilPrograma de Pós-Graduação em Endocrinología, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brasil
- Universidade Federal de Ciências da Saúde de Porto AlegreDepartamento de NutriçãoPorto AlegreRSBrasilDepartamento de Nutrição, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brasil
| | - André F. Reis
- Universidade Federal de São PauloDepartamento de MedicinaSão PauloSPBrasilUniversidade Federal de São Paulo (Unifesp), Departamento de Medicina, Disciplina de Endocrinologia, São Paulo, SP, Brasil
| | - Fernando Gerchman
- Universidade Federal do Rio Grande do SulFaculdade de MedicinaPorto AlegreRSBrasilPrograma de Pós-Graduação em Endocrinología, Faculdade de Medicina, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brasil
- Hospital de Clínicas de Porto Alegre (HCPA)Porto AlegreRSBrasilUnidade de Metabolismo, Divisão de Endocrinologia, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, RS, Brasil
| |
Collapse
|
19
|
Zhang T, Ma Y, Wang H, Loor JJ, Xu H, Shi H, Luo J. Trans10, cis12 conjugated linoleic acid increases triacylglycerol accumulation in goat mammary epithelial cells in vitro. Anim Sci J 2017; 89:432-440. [DOI: 10.1111/asj.12935] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 08/25/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Tianying Zhang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Yue Ma
- Shaanxi Key Laboratory of Molecular Biology for Agriculture; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Hui Wang
- Shaanxi Key Laboratory of Molecular Biology for Agriculture; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics; Department of Animal Sciences and Division of Nutritional Sciences; University of Illinois; Urbana IL USA
| | - Huifen Xu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Huaiping Shi
- Shaanxi Key Laboratory of Molecular Biology for Agriculture; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture; College of Animal Science and Technology; Northwest A&F University; Yangling Shaanxi China
| |
Collapse
|
20
|
Shen W, McIntosh MK. Nutrient Regulation: Conjugated Linoleic Acid's Inflammatory and Browning Properties in Adipose Tissue. Annu Rev Nutr 2017; 36:183-210. [PMID: 27431366 DOI: 10.1146/annurev-nutr-071715-050924] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Obesity is the most widespread nutritional disease in the United States. Developing effective and safe strategies to manage excess body weight is therefore of paramount importance. One potential strategy to reduce obesity is to consume conjugated linoleic acid (CLA) supplements containing isomers cis-9, trans-11 and trans-10, cis-12, or trans-10, cis-12 alone. Proposed antiobesity mechanisms of CLA include regulation of (a) adipogenesis, (b) lipid metabolism, (c) inflammation, (d) adipocyte apoptosis, (e) browning or beiging of adipose tissue, and (f) energy metabolism. However, causality of CLA-mediated responses to body fat loss, particularly the linkage between inflammation, thermogenesis, and energy metabolism, is unclear. This review examines whether CLA's antiobesity properties are due to inflammatory signaling and considers CLA's linkage with lipogenesis, lipolysis, thermogenesis, and browning of white and brown adipose tissue. We propose a series of questions and studies to interrogate the role of the sympathetic nervous system in mediating CLA's antiobesity properties.
Collapse
Affiliation(s)
- Wan Shen
- Department of Nutrition, The University of North Carolina at Greensboro, Greensboro, North Carolina 27402; ,
| | - Michael K McIntosh
- Department of Nutrition, The University of North Carolina at Greensboro, Greensboro, North Carolina 27402; ,
| |
Collapse
|
21
|
Xia YC, Radwan A, Keenan CR, Langenbach SY, Li M, Radojicic D, Londrigan SL, Gualano RC, Stewart AG. Glucocorticoid Insensitivity in Virally Infected Airway Epithelial Cells Is Dependent on Transforming Growth Factor-β Activity. PLoS Pathog 2017; 13:e1006138. [PMID: 28046097 PMCID: PMC5234851 DOI: 10.1371/journal.ppat.1006138] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 01/13/2017] [Accepted: 12/19/2016] [Indexed: 12/15/2022] Open
Abstract
Asthma and chronic obstructive pulmonary disease (COPD) exacerbations are commonly associated with respiratory syncytial virus (RSV), rhinovirus (RV) and influenza A virus (IAV) infection. The ensuing airway inflammation is resistant to the anti-inflammatory actions of glucocorticoids (GCs). Viral infection elicits transforming growth factor-β (TGF-β) activity, a growth factor we have previously shown to impair GC action in human airway epithelial cells through the activation of activin-like kinase 5 (ALK5), the type 1 receptor of TGF-β. In the current study, we examine the contribution of TGF-β activity to the GC-resistance caused by viral infection. We demonstrate that viral infection of human bronchial epithelial cells with RSV, RV or IAV impairs GC anti-inflammatory action. Poly(I:C), a synthetic analog of double-stranded RNA, also impairs GC activity. Both viral infection and poly(I:C) increase TGF-β expression and activity. Importantly, the GC impairment was attenuated by the selective ALK5 (TGFβRI) inhibitor, SB431542 and prevented by the therapeutic agent, tranilast, which reduced TGF-β activity associated with viral infection. This study shows for the first time that viral-induced glucocorticoid-insensitivity is partially mediated by activation of endogenous TGF-β. In this study, we investigate how respiratory viral infection interferes with the anti-inflammatory actions of glucocorticoid (GC) drugs, which are a highly effective group of anti-inflammatory agents widely used in the treatment of chronic inflammatory airway diseases, including asthma and chronic obstructive pulmonary disease (COPD). Exacerbations of both asthma (“asthma attacks”) and COPD are often caused by viral infection, which does not respond well to GC therapy. Patients are often hospitalized placing a large burden on healthcare systems around the world, with the young, elderly, and those with a poor immune system particularly at risk. We show that viral infection of airway epithelial cells causes increased expression and activity of transforming growth factor-beta (TGF-β), which interferes with GC drug action. Importantly, we have shown for the first time that inhibiting TGF-β activity in the airways could serve as a new strategy to prevent and/or treat viral exacerbations of chronic airway diseases.
Collapse
Affiliation(s)
- Yuxiu C. Xia
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Asmaa Radwan
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Christine R. Keenan
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Shenna Y. Langenbach
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Meina Li
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Danica Radojicic
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Sarah L. Londrigan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Rosa C. Gualano
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
| | - Alastair G. Stewart
- Lung Health Research Centre, Department of Pharmacology & Therapeutics, The University of Melbourne, Parkville, Victoria, Australia
- * E-mail:
| |
Collapse
|
22
|
Vahmani P, Meadus WJ, Rolland DC, Duff P, Dugan MER. Trans10,cis15 18:2 Isolated from Beef Fat Does Not Have the Same Anti-Adipogenic Properties as Trans10,cis12–18:2 in 3T3-L1 Adipocytes. Lipids 2016; 51:1231-1239. [DOI: 10.1007/s11745-016-4192-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 08/22/2016] [Indexed: 10/21/2022]
|
23
|
Yeganeh A, Taylor CG, Tworek L, Poole J, Zahradka P. Trans-10,cis-12 conjugated linoleic acid (CLA) interferes with lipid droplet accumulation during 3T3-L1 preadipocyte differentiation. Int J Biochem Cell Biol 2016; 76:39-50. [PMID: 27131602 DOI: 10.1016/j.biocel.2016.04.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 04/14/2016] [Accepted: 04/25/2016] [Indexed: 12/22/2022]
Abstract
In this study, we hypothesize that the biologically active isomers of conjugated linoleic acid (CLA), cis-9,trans-11 (c9,t11) and trans-10,cis-12 (t10,c12) CLA, have different effects on early and late stages 3T3-L1 preadipocyte differentiation. Both c9-t11 and t10-c12CLA stimulated early stage pre-adipocyte differentiation (day 2), while t10-c12CLA inhibited late differentiation (day 8) as determined by lipid droplet numbers and both perilipin-1 levels and phosphorylation state. At day 8, the adipokines adiponectin, chemerin and adipsin were all reduced in t10-c12CLA treated cells versus control cells. Immunofluorescence microscopy showed perilipin-1 was present solely on lipid droplets on day 8 in t10-c12 treated 3T3-L1 cells, whereas preilipin-1 was also located in the perinuclear region in control and c9-t11 treated cells. The t10-c12CLA isomer also decreased levels of hormone-sensitive lipase and inhibited lipolysis. These findings indicate that the decrease in lipid droplets caused by t10-c12CLA is the result of an inhibition of lipid droplet production during adipogenesis rather than a stimulation of lipolysis. Additionally, treatment with Gö6976 blocked the effect of t10-c12CLA on perilipin-1 phosphorylation, implicating PKCα in perilipin-1 phosphorylation, and thus a regulator of triglyceride catabolism. These data are supported by evidence that t10-c12CLA activated PKCα. These are the first data to show that CLA isomers can affect lipid droplet dynamics in adipocytes through PKCα.
Collapse
Affiliation(s)
- Azadeh Yeganeh
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Canada
| | - Carla G Taylor
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada; Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Canada
| | - Leslee Tworek
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Canada
| | - Jenna Poole
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Canada
| | - Peter Zahradka
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada; Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, Canada; Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, Canada.
| |
Collapse
|
24
|
Zhao L, Yagiz Y, Xu C, Lu J, Chung S, Marshall MR. Muscadine grape seed oil as a novel source of tocotrienols to reduce adipogenesis and adipocyte inflammation. Food Funct 2016; 6:2293-302. [PMID: 26073057 DOI: 10.1039/c5fo00261c] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Tocotrienols are unsaturated forms of vitamin E previously shown to reduce adipogenesis and adipose inflammation. In this study, muscadine grape seed oil (MGSO) was identified as a novel source of tocotrienols containing significant amounts of α- and γ-tocotrienol (T3) with minor seasonal changes. The aim of this study was to assess the anti-adipogenic and anti-inflammatory potential of MGSO by using primary human adipose-derived stem cells (hASCs). Differentiating hASCs were treated with MGSO and compared with rice bran and olive oil. Accumulation of triglyceride was significantly lower in MGSO-treated hASCs than rice bran and olive oils. A tocotrienol rich fraction (TRF) from MGSO was prepared by solid phase extraction and eluted with 15% 1,4-dioxane in hexane. The MGSO-derived TRF treatment significantly reduced mRNA and protein expression that are crucial to adipogenesis (e.g., PPARγ and aP2) in hASCs. Furthermore, TRF from MGSO markedly reduced LPS-induced proinflammatory gene expression in human adipocytes and cytokine secretion to the medium (IL-6 and IL-8). Collectively, our work suggests that MGSO is a stable and reliable natural source of T3 and MGSO may constitute a new dietary strategy to attenuate obesity and its associated adipose inflammation.
Collapse
Affiliation(s)
- Lu Zhao
- Department of Food Science and Human Nutrition, University of Florida, Gainesville 32611, Florida, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Woo H, Chung MY, Kim J, Kong D, Min J, Choi HD, Choi IW, Kim IH, Noh SK, Kim BH. Conjugated Linoleic Triacylglycerols Exhibit Superior Lymphatic Absorption Than Free Conjugate Linoleic Acids and Have Antiobesity Properties. J Med Food 2016; 19:486-94. [PMID: 27081749 DOI: 10.1089/jmf.2015.3627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This study aimed to compare lymphatic absorption of conjugated linoleic acids (CLAs) in the triacylglycerol (TAG) or free fatty acid (FFA) form and to examine the antiobesity effects of different doses of CLAs in the TAG form in animals. Conjugated linoleic TAGs (containing 70.3 wt% CLAs; CLA-TAG) were prepared through lipase-catalyzed esterification of glycerol with commercial CLA mixtures (CLA-FFA). Lymphatic absorption of CLA-TAG and CLA-FFA was compared in a rat model of lymphatic cannulation. Greater amounts of cis-9,trans-11 and trans-10,cis-12 CLAs were detected in the collected lymph from a lipid emulsion containing CLA-TAG. This result suggests that CLA-TAG has greater capacity for lymphatic absorption than does CLA-FFA. The antiobesity efficacy of CLA-TAG at different doses was examined in mice with diet-induced obesity. A high-fat diet (HFD) for 12 weeks caused a significant increase in body weight and epididymal and retroperitoneal fat weights, which were significantly decreased by 2% dietary supplementation (w/w) with CLA-TAG. CLA-TAG at 2% significantly attenuated the HFD-induced upregulation of serum TAG, but led to hepatomegaly and exacerbated HFD-induced hypercholesterolemia. CLA-TAG at 1% significantly attenuated upregulation of retroperitoneal fat weight and significantly increased liver weight, which was decreased by the HFD. Nonetheless, the liver weight in group "HFD +1% CLA-TAG" was not significantly different from that of normal diet controls. CLA-TAG at 1% significantly reduced serum TAG levels and did not exacerbate HFD-induced hypercholesterolemia. Thus, 1% dietary supplementation with CLA-TAG reduces retroperitoneal fat weight without apparent hepatomegaly, a known side-effect of CLAs in mouse models of obesity.
Collapse
Affiliation(s)
- Hyunjoon Woo
- 1 Department of Food Science and Technology, Chung-Ang University , Anseong, Korea
| | - Min-Yu Chung
- 2 Korea Food Research Institute , Seongnam, Korea
| | - Juyeon Kim
- 3 Department of Food and Nutrition, Changwon National University , Changwon, Korea
| | - Daecheol Kong
- 3 Department of Food and Nutrition, Changwon National University , Changwon, Korea
| | - Jinyoung Min
- 2 Korea Food Research Institute , Seongnam, Korea
| | - Hee-Don Choi
- 2 Korea Food Research Institute , Seongnam, Korea
| | - In-Wook Choi
- 2 Korea Food Research Institute , Seongnam, Korea
| | - In-Hwan Kim
- 4 Department of Food and Nutrition, Korea University , Seoul, Korea
| | - Sang K Noh
- 3 Department of Food and Nutrition, Changwon National University , Changwon, Korea
| | - Byung Hee Kim
- 5 Department of Food and Nutrition, Sookmyung Women's University , Seoul, Korea
| |
Collapse
|
26
|
Trans10, cis12 conjugated linoleic acid inhibits 3T3-L1 adipocyte adipogenesis by elevating β-catenin levels. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:363-70. [DOI: 10.1016/j.bbalip.2016.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 12/22/2015] [Accepted: 01/08/2016] [Indexed: 12/30/2022]
|
27
|
Dietary docosahexaenoic acid reverses nonalcoholic steatohepatitis and fibrosis caused by conjugated linoleic acid supplementation in mice. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.11.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
28
|
Low level of trans-10, cis-12 conjugated linoleic acid decreases adiposity and increases browning independent of inflammatory signaling in overweight Sv129 mice. J Nutr Biochem 2015; 26:616-25. [PMID: 25801353 DOI: 10.1016/j.jnutbio.2014.12.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 11/06/2014] [Accepted: 12/19/2014] [Indexed: 01/04/2023]
Abstract
The objective of this study was to determine the extent to which a low level of trans-10, cis-12 (10,12) conjugated linoleic acid (CLA) decreases adiposity and increases browning in overweight mice, its dependence on inflammatory signaling and potential synergistic effects of daily exercise. Young, Sv129 male mice were fed a high-fat diet for 5 weeks to make them fat and glucose intolerant and then switch them to a low-fat diet with or without 0.1% 10,12 CLA, sodium salicylate or exercise for another 7 weeks. 10,12 CLA decreased white adipose tissue (WAT) and brown adipose tissue mass, and increased the messenger RNA and protein levels, and activities of enzymes associated with thermogenesis or fatty acid oxidation in WAT. Mice fed 10,12 CLA had lower body temperatures compared to controls during cold exposure, which coincided with decreased adiposity. Although sodium salicylate decreased 10,12 CLA-mediated increases in markers of inflammation in WAT, it did not affect other outcomes. Exercise had no further effect on the outcomes measured. Collectively, these data indicate that 10,12 CLA-mediated reduction of adiposity is independent of inflammatory signaling, and possibly due to up-regulation of fatty acid oxidation and heat production in order to regulate body temperature. Although this low level of 10,12 CLA reduced adiposity in overweight mice, hepatomegaly and inflammation are major health concerns.
Collapse
|
29
|
Yuan G, Chen X, Li D. Modulation of peroxisome proliferator-activated receptor gamma (PPAR γ) by conjugated fatty acid in obesity and inflammatory bowel disease. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:1883-1895. [PMID: 25634802 DOI: 10.1021/jf505050c] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Conjugated fatty acids including conjugated linoleic acid (CLA) and conjugated linolenic acid (CLNA) have drawn significant attention for their variety of biologically beneficial effects. Evidence suggested that CLA and CLNA could play physiological roles by regulating the expression and activity of PPAR γ. This review summarizes the current understanding of evidence of the role of CLA (cis-9,trans-11 CLA and trans-10,cis-12 CLA) and CLNA (punicic acid and α-eleostearic acid) in modulating the expression or activity of PPAR γ that could in turn be employed as complementary treatment for obesity and inflammatory bowel disease.
Collapse
Affiliation(s)
- Gaofeng Yuan
- Zhejiang Provincial Key Laboratory of Health Risk Factors for Seafood, Zhejiang Ocean University , Zhoushan 316022, China
| | | | | |
Collapse
|
30
|
Abstract
Protein arrays have shown potential applications in cancer research. After several decades of research, it has become evident that many cytokines are central to the development of cancer and its treatment. Cytokine antibody arrays that have been designed to simultaneously detect multiple cytokines are not only available, but show a diversity of applications in the study of many diseases in addition to cancer. This review will focus on the implementation of cytokine antibody arrays in many aspects of cancer research, such as biomarker discovery, molecular mechanisms of cancer development, preclinical studies and the effects of cancer compounds.
Collapse
Affiliation(s)
- Ruo-Pan Huang
- Emory University School of Medicine, Department of Gynecology & Obstetrics, Atlanta, GA 30322, USA.
| |
Collapse
|
31
|
|
32
|
Jia B, Wu G, Fu X, Mo X, Du M, Hou Y, Zhu S. trans
-10, cis
-12 conjugated linoleic acid enhances in vitro maturation of porcine oocytes. Mol Reprod Dev 2013; 81:20-30. [DOI: 10.1002/mrd.22273] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 10/19/2013] [Indexed: 12/31/2022]
Affiliation(s)
- Baoyu Jia
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| | - Guoquan Wu
- Yunnan Animal Science and Veterinary Institute; Kunming Yunnan People's Republic of China
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| | - Xianhong Mo
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences; China Agricultural University; Beijing People's Republic of China
| | - Ming Du
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| | - Yunpeng Hou
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences; China Agricultural University; Beijing People's Republic of China
| | - Shien Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology; China Agricultural University; Beijing People's Republic of China
| |
Collapse
|
33
|
Thomas G, Betters JL, Lord CC, Brown AL, Marshall S, Ferguson D, Sawyer J, Davis MA, Melchior JT, Blume LC, Howlett AC, Ivanova PT, Milne SB, Myers DS, Mrak I, Leber V, Heier C, Taschler U, Blankman JL, Cravatt BF, Lee RG, Crooke RM, Graham MJ, Zimmermann R, Brown HA, Brown JM. The serine hydrolase ABHD6 Is a critical regulator of the metabolic syndrome. Cell Rep 2013; 5:508-20. [PMID: 24095738 DOI: 10.1016/j.celrep.2013.08.047] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 07/25/2013] [Accepted: 08/29/2013] [Indexed: 01/31/2023] Open
Abstract
The serine hydrolase α/β hydrolase domain 6 (ABHD6) has recently been implicated as a key lipase for the endocannabinoid 2-arachidonylglycerol (2-AG) in the brain. However, the biochemical and physiological function for ABHD6 outside of the central nervous system has not been established. To address this, we utilized targeted antisense oligonucleotides (ASOs) to selectively knock down ABHD6 in peripheral tissues in order to identify in vivo substrates and understand ABHD6's role in energy metabolism. Here, we show that selective knockdown of ABHD6 in metabolic tissues protects mice from high-fat-diet-induced obesity, hepatic steatosis, and systemic insulin resistance. Using combined in vivo lipidomic identification and in vitro enzymology approaches, we show that ABHD6 can hydrolyze several lipid substrates, positioning ABHD6 at the interface of glycerophospholipid metabolism and lipid signal transduction. Collectively, these data suggest that ABHD6 inhibitors may serve as therapeutics for obesity, nonalcoholic fatty liver disease, and type II diabetes.
Collapse
Affiliation(s)
- Gwynneth Thomas
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Jenna L Betters
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Caleb C Lord
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Amanda L Brown
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,New Affiliation: Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland OH 44195, USA
| | - Stephanie Marshall
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,New Affiliation: Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland OH 44195, USA
| | - Daniel Ferguson
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,New Affiliation: Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland OH 44195, USA
| | - Janet Sawyer
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Matthew A Davis
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - John T Melchior
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Lawrence C Blume
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Allyn C Howlett
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA
| | - Pavlina T Ivanova
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Stephen B Milne
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - David S Myers
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Irina Mrak
- Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Vera Leber
- Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Christoph Heier
- Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Ulrike Taschler
- Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - Jacqueline L Blankman
- Deparment of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Benjamin F Cravatt
- Deparment of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Richard G Lee
- Cardiovascular Group, Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA 92010 USA
| | - Rosanne M Crooke
- Cardiovascular Group, Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA 92010 USA
| | - Mark J Graham
- Cardiovascular Group, Antisense Drug Discovery, Isis Pharmaceuticals, Inc., Carlsbad, CA 92010 USA
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, A-8010 Graz, Austria
| | - H Alex Brown
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - J Mark Brown
- Department of Pathology, Section on Lipid Sciences, Wake Forest University School of Medicine, Winston-Salem, NC 27157, USA.,New Affiliation: Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, Cleveland OH 44195, USA
| |
Collapse
|
34
|
den Hartigh LJ, Han CY, Wang S, Omer M, Chait A. 10E,12Z-conjugated linoleic acid impairs adipocyte triglyceride storage by enhancing fatty acid oxidation, lipolysis, and mitochondrial reactive oxygen species. J Lipid Res 2013; 54:2964-78. [PMID: 23956445 DOI: 10.1194/jlr.m035188] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Conjugated linoleic acid (CLA) is a naturally occurring dietary trans fatty acid found in food from ruminant sources. One specific CLA isomer, 10E,12Z-CLA, has been associated with health benefits, such as reduced adiposity, while simultaneously promoting deleterious effects, such as systemic inflammation, insulin resistance, and dyslipidemia. The precise mechanisms by which 10E,12Z-CLA exerts these effects remain unknown. Despite potential health consequences, CLA continues to be advertised as a natural weight loss supplement, warranting further studies on its effects on lipid metabolism. We hypothesized that 10E,12Z-CLA impairs lipid storage in adipose tissue by altering the lipid metabolism of white adipocytes. We demonstrate that 10E,12Z-CLA reduced triglyceride storage due to enhanced fatty acid oxidation and lipolysis, coupled with diminished glucose uptake and utilization in cultured adipocytes. This switch to lipid utilization was accompanied by a potent proinflammatory response, including the generation of cytokines, monocyte chemotactic factors, and mitochondrial superoxide. Disrupting fatty acid oxidation restored glucose utilization and attenuated the inflammatory response to 10E,12Z-CLA, suggesting that fatty acid oxidation is critical in promoting this phenotype. With further investigation into the biochemical pathways involved in adipocyte responses to 10E,12Z-CLA, we can discern more information about its safety and efficacy in promoting weight loss.
Collapse
Affiliation(s)
- Laura J den Hartigh
- Diabetes Obesity Center for Excellence and the Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington, Seattle, WA 98109
| | | | | | | | | |
Collapse
|
35
|
Kim Y, Kelly OJ, Ilich JZ. Synergism of α-linolenic acid, conjugated linoleic acid and calcium in decreasing adipocyte and increasing osteoblast cell growth. Lipids 2013; 48:787-802. [PMID: 23757205 DOI: 10.1007/s11745-013-3803-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 05/07/2013] [Indexed: 11/30/2022]
Abstract
Whole fat milk and dairy products (although providing more energy compared to low- or non-fat products), are good sources of α-linolenic acid (ALA), conjugated linoleic acid (CLA) and calcium, which may be favorable in modulating bone and adipose tissue metabolism. We examined individual and/or synergistic effects of ALA, CLA and calcium (at levels similar to those in whole milk/dairy products) in regulating bone and adipose cell growth. ST2 stromal, MC3T3-L1 adipocyte-like and MC3T3-E1 osteoblast-like cells were treated with: (a) linoleic acid (LNA):ALA ratios = 1-5:1; (b) individual/combined 80-90 % c9, t11 (9,11) and 5-10 % t10, c12 (10,12) CLA isomers; (c) 0.5-3.0 mM calcium; (d) combinations of (a), (b), (c); and (e) control. Local mediators, including eicosanoids and growth factors, were measured. (a) The optimal effect was found at the 4:1 LNA:ALA ratio where insulin-like growth factor-1 (IGF-1) and IGF binding protein-3 (IGFBP-3) production was the lowest in MC3T3-L1 cells. (b) All CLA isomer blends decreased MC3T3-L1 and increased MC3T3-E1 cell differentiation. (c) 1.5-2.5 mM calcium increased ST2 and MC3T3-E1, and decreased MC3T3-L1 cell proliferation. (d) Combination of 4:1 LNA:ALA + 90:10 % CLA + 2.0 mM calcium lowered MC3T3-L1 and increased MC3T3-E1 cell differentiation. Overall, the optimal LNA:ALA ratio to enhance osteoblastogenesis and inhibit adipogenesis was 4:1. This effect was enhanced by 90:10 % CLA + 2.0 mM calcium, indicating possible synergism of these dietary factors in promoting osteoblast and inhibiting adipocyte differentiation in cell cultures.
Collapse
Affiliation(s)
- Youjin Kim
- Bayer CropScience Ltd., Gangnam-gu, Seoul 135-979, South Korea
| | | | | |
Collapse
|
36
|
Shen W, Martinez K, Chuang CC, McIntosh M. The phospholipase C inhibitor U73122 attenuates trans-10, cis-12 conjugated linoleic acid-mediated inflammatory signaling and insulin resistance in human adipocytes. J Nutr 2013; 143:584-90. [PMID: 23468551 PMCID: PMC3738231 DOI: 10.3945/jn.112.173161] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We have demonstrated that trans-10, cis-12 conjugated linoleic acid (18:2t10,c12)-mediated delipidation of human adipocytes was dependent on increased intracellular calcium and activation of inflammatory signaling in human primary adipocytes. These data are consistent with the actions of diacylglycerol and inositol triphosphate derived from phospholipase C (PLC)-dependent cell signaling. To test the hypothesis that PLC was an upstream activator of these cellular responses to 18:2t10,c12, primary cultures of human adipocytes were pretreated with 1-[6-((17β-3-methoxyestra-1,3,5 (10)-trien-17-yl)amino)hexyl]-1H-pyrrole-2,5-dione (U73122), a universal PLC inhibitor, followed by 18:2t10,c12 treatment. U73122 attenuated 18:2t10,c12-mediated insulin resistance within 48 h and suppression of the mRNA levels of peroxisome proliferator-activated receptor (PPAR)γ, insulin-stimulated glucose transporter-4, acetyl-CoA carboxylase-1, and stearoyl-CoA desaturase-1, and the protein levels of PPARγ within 18-24 h. U73122 inhibited 18:2t10,c12-mediated induction of the inflammatory-related genes calcium/calmodulin-dependent protein kinase-β, cyclooxygenase-2, monocyte chemoattractant protein-1, interleukin (IL)-6, and IL-8, secretion of IL-6 and IL-8, and the activation of extracellular signal-related kinase, c-Jun N-terminal kinase, and c-Jun within 18-24 h. Moreover, 18:2t10,c12 increased the mRNA levels of heat shock proteins within 6-24 h and intracellular calcium concentrations within 3 min, which were inhibited by U73122. Lastly, 18:2t10,c12 increased the abundance of PLCγ1 in the plasma membrane within 3 min. Taken together, these data suggest that PLC plays an important role in 18:2t10,c12-mediated activation of intracellular calcium accumulation, inflammatory signaling, delipidation, and insulin resistance in human primary adipocytes.
Collapse
|
37
|
Effects of conjugated linoleic acid on growth, non-specific immunity, antioxidant capacity, lipid deposition and related gene expression in juvenile large yellow croaker (Larmichthys crocea) fed soyabean oil-based diets. Br J Nutr 2013; 110:1220-32. [DOI: 10.1017/s0007114513000378] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The effects of conjugated linoleic acid (CLA) on growth performance, non-specific immunity, antioxidant capacity, lipid deposition and related gene expression were investigated in the large yellow croaker (Larmichthys crocea). Fish (7·56 (sem 0·60) g) were fed soyabean oil-based diets with graded levels of CLA (0, 0·42, 0·83, 1·70 %) for 70 d. Quantitative PCR was used to assess the effects of CLA on the transcription of inflammation- and fatty acid oxidation-related genes. Growth in fish fed the diet with 0·42 % CLA was significantly higher. Also, phagocytic index and respiratory burst activity were significantly higher in fish fed the diets containing 0·42 and 0·83 % CLA, respectively. Hepatic total antioxidative capacity and catalase activities increased significantly when CLA increased from 0 to 0·83 %, and then decreased with further increase of CLA. However, hepatic malondialdehyde content decreased significantly as dietary CLA increased. Lipid concentration in the whole body and muscle increased significantly with increasing dietary CLA. Transcription of genes related to inflammation (cyclo-oxygenase-2 and IL-β) in the liver and kidney and fatty acid oxidation (carnitine palmitoyl transferase I and acyl CoA oxidase) in the kidney decreased significantly as dietary CLA increased. PPARα and acyl CoA oxidase expression in the liver decreased significantly as CLA increased from 0·42 to 1·70 %. These results strongly suggest that dietary CLA could significantly affect growth performance, non-specific immunity, antioxidant capacity, lipid deposition and transcription of inflammation- and fatty acid oxidation-related genes of the large yellow croaker. This may contribute to our understanding of the mechanisms related to the physiological effects of dietary CLA in fish.
Collapse
|
38
|
Martinez K, Shyamasundar S, Kennedy A, Chuang CC, Marsh A, Kincaid J, Reid T, McIntosh M. Diacylglycerol kinase inhibitor R59022 attenuates conjugated linoleic acid-mediated inflammation in human adipocytes. J Lipid Res 2013; 54:662-670. [PMID: 23264678 PMCID: PMC3617941 DOI: 10.1194/jlr.m031211] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 12/17/2012] [Indexed: 11/20/2022] Open
Abstract
Diacylglycerol kinases (DGK) convert diacylglycerol to phosphatidic acid, which has been reported to stimulate calcium release from the endoplasmic reticulum. Based on our published data showing that trans-10, cis-12 conjugated linoleic acid (t10,c12 CLA)-mediated intracellular calcium accumulation is linked to inflammation and insulin resistance, we hypothesized that inhibiting DGKs with R59022 would prevent t10,c12 CLA-mediated inflammatory signaling and insulin resistance in human adipocytes. Consistent with our hypothesis, R59022 attenuated t10,c12 CLA-mediated i) increased gene expression and protein secretion of interleukin (IL)-8, IL-6, and monocyte chemoattractant protein-1 (MCP-1); ii) increased activation of extracellular signal-related kinase (ERK), cJun-NH2-terminal kinase (JNK), and cJun; iii) increased intracellular calcium levels; iv) suppressed mRNA or protein levels of peroxisome proliferator activated receptor γ, adiponectin, and insulin-dependent glucose transporter 4; and v) decreased fatty acid and glucose uptake and triglyceride content. DGKη was targeted for investigation based on our findings that i) DGKη was highly expressed in primary human adipocytes and time-dependently induced by t10,c12 CLA and that ii) t10,c12 CLA-induced DGKη expression was dose-dependently decreased with R59022. Small interfering RNA (siRNA) targeting DGKη decreased t10,c12 CLA-induced DGKη, IL-8, and MCP-1 gene expression, as well as activation of JNK and cJun. Taken together, these data suggest that DGKs mediate, in part, t10,c12 CLA-induced inflammatory signaling in primary human adipocytes.
Collapse
Affiliation(s)
- Kristina Martinez
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC
| | - Shruthi Shyamasundar
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC
| | - Arion Kennedy
- Department of Molecular Physiology, Vanderbilt University Medical Center, Nashville, TN
| | - Chia Chi Chuang
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC
| | - Angel Marsh
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC
| | - Jennifer Kincaid
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC
| | - Tanya Reid
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC
| | - Michael McIntosh
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC
| |
Collapse
|
39
|
Shen W, Chuang CC, Martinez K, Reid T, Brown JM, Xi L, Hixson L, Hopkins R, Starnes J, McIntosh M. Conjugated linoleic acid reduces adiposity and increases markers of browning and inflammation in white adipose tissue of mice. J Lipid Res 2013; 54:909-22. [PMID: 23401602 DOI: 10.1194/jlr.m030924] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The objective of this study was to examine the mechanism by which conjugated linoleic acid (CLA) reduces body fat. Young male mice were fed three combinations of fatty acids at three doses (0.06%, 0.2%, and 0.6%, w/w) incorporated into AIN76 diets for 7 weeks. The types of fatty acids were linoleic acid (control), an equal mixture of trans-10, cis-12 (10,12) CLA plus linoleic acid, and an equal isomer mixture of 10,12 plus cis-9, trans-11 (9,11) CLA. Mice receiving the 0.2% and 0.6% dose of 10,12 CLA plus linoleic acid or the CLA isomer mixture had decreased white adipose tissue (WAT) and brown adipose tissue (BAT) mass and increased incorporation of CLA isomers in epididymal WAT and liver. Notably, in mice receiving 0.2% of both CLA treatments, the mRNA levels of genes associated with browning, including uncoupling protein 1 (UCP1), UCP1 protein levels, and cytochrome c oxidase activity, were increased in epididymal WAT. CLA-induced browning in WAT was accompanied by increases in mRNA levels of markers of inflammation. Muscle cytochrome c oxidase activity and BAT UCP1 protein levels were not affected by CLA treatment. These data suggest a linkage between decreased adiposity, browning in WAT, and low-grade inflammation due to consumption of 10,12 CLA.
Collapse
Affiliation(s)
- Wan Shen
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Zhou YJ, Zhou H, Li Y, Song YL. NOD1 activation induces innate immune responses and insulin resistance in human adipocytes. DIABETES & METABOLISM 2012. [PMID: 23182460 DOI: 10.1016/j.diabet.2012.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
AIMS The innate immune-receptor nucleotide oligomerization domain (NOD) protein recognizes intracellular bacterial peptidoglycan. Activation of the innate immune system contributes to the development and progression of insulin resistance. The present study aimed to determine the presence of NOD1 and NOD2 in human adipose cells as well as to assess their functionality. METHODS Subcutaneous abdominal fat from obese subjects was biopsied and characterized for NOD expression using quantitative real-time PCR (qPCR). Human adipocytes were stimulated with iE-DAP (NOD1-specific ligand), and NOD1, proinflammatory cytokine production and nuclear factor (NF)-κB activation were quantified using qPCR, enzyme-linked immunosorbent assay (Elisa) and luciferase assay. Insulin-stimulated glucose uptake was determined by measuring 2-deoxy-D-[(3)H] glucose uptake. Expression and phosphorylation of IRS-1, Akt and JNK were evaluated using Western blotting. RESULTS NOD1/NOD2 mRNA expression was induced during adipocyte differentiation and enhanced in human adipose depots. Stimulation of isolated human adipocytes with iE-DAP induced NF-κB p65 nuclear translocation and a marked increase in proinflammatory cytokine production, including MCP-1, IL-6 and IL-8. NOD1 activation weakened insulin signal transduction as revealed by increased JNK and IRS-1 Ser307 phosphorylation, inhibited IRS-1 tyrosine phosphorylation, and reduced insulin-induced phosphorylation of Akt on Ser473 and Thr308 in human adipocytes. Moreover, NOD1 activation reduced insulin-induced glucose uptake, leading to insulin resistance. CONCLUSION These results suggest that NOD1 signaling could be one of the links between innate immunity and insulin resistance in human adipocytes. This study provides supporting evidence for NOD1 protein as a component of innate immunity involved in insulin resistance.
Collapse
Affiliation(s)
- Y-J Zhou
- Department of Endocrinology and Metabolism, Fourth Affiliated Hospital, China Medical University, 110032 Shenyang, PR China.
| | | | | | | |
Collapse
|
41
|
Reardon M, Gobern S, Martinez K, Shen W, Reid T, McIntosh M. Oleic acid attenuates trans-10,cis-12 conjugated linoleic acid-mediated inflammatory gene expression in human adipocytes. Lipids 2012; 47:1043-51. [PMID: 22941440 DOI: 10.1007/s11745-012-3711-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 08/13/2012] [Indexed: 11/25/2022]
Abstract
The weight loss supplement conjugated linoleic acid (CLA) consists of an equal mixture of trans-10,cis-12 (10,12) and cis-9,trans-11 (9,11) isomers. However, high levels of mixed CLA isomers, or the 10,12 isomer, causes chronic inflammation, lipodystrophy, or insulin resistance. We previously demonstrated that 10,12 CLA decreases de novo lipid synthesis along with the abundance and activity of stearoyl-CoA desaturase (SCD)-1, a δ-9 desaturase essential for the synthesis of monounsaturated fatty acids (MUFA). Thus, we hypothesized that the 10,12 CLA-mediated decrease in SCD-1, with the subsequent decrease in MUFA, was responsible for the observed effects. To test this hypothesis, 10,12 CLA-treated human adipocytes were supplemented with oleic acid for 12 h to 7 days, and inflammatory gene expression, insulin-stimulated glucose uptake, and lipid content were measured. Oleic acid reduced inflammatory gene expression in a dose-dependent manner, and restored the lipid content of 10,12 CLA-treated adipocytes without improving insulin-stimulated glucose uptake. In contrast, supplementation with stearic acid, a substrate for SCD-1, or 9,11 CLA did not prevent inflammatory gene expression by 10,12 CLA. Notably, 10,12 CLA impacted the expression of several G-protein coupled receptors that was attenuated by oleic acid. Collectively, these data show that oleic acid attenuates 10,12 CLA-induced inflammatory gene expression and lipid content, possibly by alleviating cell stress caused by the inhibition of MUFA needed for phospholipid and neutral lipid synthesis.
Collapse
Affiliation(s)
- Meaghan Reardon
- Department of Nutrition, University of North Carolina at Greensboro, Greensboro, NC 27402-6170, USA.
| | | | | | | | | | | |
Collapse
|
42
|
Acute exposure of L6 myotubes to cis-9, trans-11 and trans-10, cis-12 conjugated linoleic acid isomers stimulates glucose uptake by modulating Ca2+/calmodulin-dependent protein kinase II. Int J Biochem Cell Biol 2012; 44:1321-30. [DOI: 10.1016/j.biocel.2012.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2012] [Revised: 05/02/2012] [Accepted: 05/08/2012] [Indexed: 01/28/2023]
|
43
|
Prophylactic effect of α-linolenic acid and α-eleostearic acid against MeHg induced oxidative stress, DNA damage and structural changes in RBC membrane. Food Chem Toxicol 2012; 50:2811-8. [DOI: 10.1016/j.fct.2012.05.038] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 05/21/2012] [Accepted: 05/23/2012] [Indexed: 12/31/2022]
|
44
|
Mohankumar SK, Taylor CG, Siemens L, Zahradka P. Activation of phosphatidylinositol-3 kinase, AMP-activated kinase and Akt substrate-160 kDa by trans-10, cis-12 conjugated linoleic acid mediates skeletal muscle glucose uptake. J Nutr Biochem 2012; 24:445-56. [PMID: 22704782 DOI: 10.1016/j.jnutbio.2012.01.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Revised: 11/09/2011] [Accepted: 01/06/2012] [Indexed: 10/28/2022]
Abstract
Conjugated linoleic acid (CLA), a dietary lipid, has been proposed as an antidiabetic agent. However, studies specifically addressing the molecular dynamics of CLA on skeletal muscle glucose transport and differences between the key isomers are limited. We demonstrate that acute exposure of L6 myotubes to cis-9, trans-11 (c9,t11) and trans-10, cis-12 (t10,c12) CLA isomers mimics insulin action by stimulating glucose uptake and glucose transporter-4 (GLUT4) trafficking. Both c9,t10-CLA and t10,c12-CLA stimulate the phosphorylation of phosphatidylinositol 3-kinase (PI3-kinase) p85 subunit and Akt substrate-160 kDa (AS160), while showing isomer-specific effects on AMP-activated protein kinase (AMPK). CLA isomers showed synergistic effects with the AMPK activator, 5-aminoimidazole-4-carboxamide-1-β-d-ribonucleoside (AICAR). Blocking PI3-kinase and AMPK prevented the stimulatory effects of t10,c12-CLA on AS160 phosphorylation and glucose uptake, indicating that this isomer acts via a PI3-kinase and AMPK-dependent mechanism, whereas the mechanism of c9,t11-CLA remains unclear. Intriguingly, CLA isomers sensitized insulin-Akt-responsive glucose uptake and prevented high insulin-induced Akt desensitisation. Together, these results establish that CLA exhibits isomer-specific effects on GLUT4 trafficking and the increase in glucose uptake induced by CLA treatment of L6 myotubes occurs via pathways that are distinctive from those utilised by insulin.
Collapse
|
45
|
Belda BJ, Thompson JT, Sinha R, Prabhu KS, Vanden Heuvel JP. The dietary fatty acid 10E12Z-CLA induces epiregulin expression through COX-2 dependent PGF(2α) synthesis in adipocytes. Prostaglandins Other Lipid Mediat 2012; 99:30-7. [PMID: 22583689 DOI: 10.1016/j.prostaglandins.2012.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 05/01/2012] [Accepted: 05/02/2012] [Indexed: 02/06/2023]
Abstract
Conjugated linoleic acids (CLAs) are a group of dietary fatty acids that are widely marketed as weight loss supplements. The isomer responsible for this effect is the trans-10, cis-12 CLA (10E12Z-CLA) isomer. 10E12Z-CLA treatment during differentiation of 3T3-L1 adipocytes induces expression of prostaglandin-endoperoxide synthase-2 (Cyclooxygenase-2; COX-2). This work demonstrates that COX-2 is also induced in fully differentiated 3T3-L1 adipocytes after a single treatment of 10E12Z-CLA at both the mRNA (20-40 fold) and protein level (7 fold). Furthermore, prostaglandin (PG)F(2α), but not PGE(2), is significantly increased 10 fold. In female BALB/c mice fed 0.5% 10E12Z-CLA for 10 days, COX-2 was induced in uterine adipose (2 fold). In vitro, pharmacological COX-2 inhibition did not block the effect of 10E12Z-CLA on adipocyte-specific gene expression although PGF(2α) was dose-dependently decreased. These studies demonstrate that PGF(2α) was not by itself responsible for the reduction in adipocyte character due to 10E12Z-CLA treatment. However, PGF(2α), either exogenously or endogenously in response to 10E12Z-CLA, increased the expression of the potent mitogen and epidermal growth factor (EGF) receptor (EGFR) ligand epiregulin in 3T3-L1 adipocytes. Blocking PGF(2α) signaling with the PGF(2α) receptor (FP) antagonist AL-8810 returned epiregulin mRNA levels back to baseline. Although this pathway is not directly responsible for adipocyte dependent gene expression, these results suggest that this signaling pathway may still have broad effect on the adipocyte and surrounding cells.
Collapse
Affiliation(s)
- Benjamin J Belda
- The Center for Molecular Toxicology and Carcinogenesis and The Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, United States
| | | | | | | | | |
Collapse
|
46
|
10E12Z CLA alters adipocyte differentiation and adipocyte cytokine expression and induces macrophage proliferation. J Nutr Biochem 2012; 23:510-8. [DOI: 10.1016/j.jnutbio.2011.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Revised: 02/16/2011] [Accepted: 02/17/2011] [Indexed: 12/26/2022]
|
47
|
Chou YC, Su HM, Lai TW, Chyuan JH, Chao PM. cis-9, trans-11, trans-13-Conjugated linolenic acid induces apoptosis and sustained ERK phosphorylation in 3T3-L1 preadipocytes. Nutrition 2012; 28:803-11. [PMID: 22465903 DOI: 10.1016/j.nut.2011.11.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 11/11/2011] [Accepted: 11/16/2011] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to investigate the adipogenic effect of cis-9, trans-11, trans-13-conjugated linolenic acid (c9,t11,t13-CLN), a fatty acid naturally present in bitter melon. METHODS The 3T3-L1 murine preadipocyte cell line was used to test the effect of saponifiables from whole bitter melon and of commercially prepared pure c9,t11,t13-CLN on adipocyte differentiation. The effect of c9,t11,t13-CLN on 3T3-L1 cell viability was also tested at proliferation, mitotic clonal expansion, and terminal differentiation stages. RESULTS Compared to the free fatty acid control mixture, the proadipogenic effect on 3T3-L1 was less potent using saponifiables obtained from bitter melon. C9,t11,t13-CLN, unlike its non-conjugated counterpart linolenic acid (LN) or other common fatty acids such as oleic acid or linoleic acid, exerted no proadipogenic effect on 3T3-L1. In contrast to LN displaying no cytotoxic effect at a concentration ≤100 μM, c9,t11,t13-CLN caused a dose-dependent reduction in the viability of pre- and postconfluent preadipocytes associated with apoptosis. Sustained ERK/MAPK activation, accompanied by increased peroxisome proliferator-activated receptor γ phosphorylation, was seen in c9,t11,t13-CLN-treated cells at initiation of differentiation. CONCLUSION C9,t11,t13-CLN is less adipogenic for 3T3-L1 cells than LN and this is partly due to its apoptotic effect on proliferating preadipocytes and to the sustained ERK phosphorylation seen during mitotic clonal expansion.
Collapse
Affiliation(s)
- Yi-Chun Chou
- Institute of Microbiology and Biochemistry, College of Life Science, National Taiwan University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
48
|
Go G, Wu G, Silvey DT, Choi S, Li X, Smith SB. Lipid metabolism in pigs fed supplemental conjugated linoleic acid and/or dietary arginine. Amino Acids 2012; 43:1713-26. [PMID: 22383090 DOI: 10.1007/s00726-012-1255-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 02/16/2012] [Indexed: 12/14/2022]
Abstract
We proposed that the combination of conjugated linoleic acid (CLA) and arginine would decrease adiposity by depressing lipid synthesis in liver and adipose tissues of growing pigs. Pigs were allotted to treatments in a 2 × 2 factorial design with two lipids (CLA or canola oil) and two amino acids [L-arginine or L-alanine (isonitrogenous control)]; supplements were provided from 80 to 110 kg body weight (approximately 4 weeks). Treatment groups (n = 4) were: control (2.05% L-alanine plus 1% canola oil); CLA (2.05% L-alanine plus 1% CLA); arginine (1.0% L-arginine plus 1.0% canola oil); arginine plus CLA (1.0% arginine plus 1.0% CLA). Arginine increased backfat thickness (P = 0.07) in the absence or presence of CLA, and arginine supplementation increased subcutaneous and retroperitoneal adipocyte volume, especially in combination with dietary CLA (interaction P = 0.001). Arginine increased palmitate incorporation into total lipids by over 60% in liver (P = 0.07). Dietary CLA increased palmitate incorporation into lipids in longissimus muscle by over 100% (P = 0.01), and CLA increased longissimus muscle lipid by nearly 20%. CLA increased glucose oxidation to CO(2) by over 80% in retroperitoneal and subcutaneous adipose tissues (P = 0.04), and doubled palmitate oxidation to CO(2) in intestinal duodenal mucosal cells (P = 0.07). Arginine supplementation decreased muscle pH at 45 min postmortem (P = 0.001), indicating elevated early postmortem glycolysis, and CLA and arginine independently increased PGC-1α gene expression in longissimus muscle. CLA but not arginine depressed mTOR gene expression in intestinal duodenal mucosal cells. CLA decreased serum insulin by 50% (P = 0.02) but increased serum triacylglycerols by over 40%. CLA supplementation increased (P ≤ 0.01) total saturated fatty acids in liver and adipose tissue. In conclusion, neither CLA nor arginine depressed tissue lipid synthesis in growing/finishing pigs, and in fact dietary CLA promoted elevated intramuscular lipid and arginine increased carcass adiposity.
Collapse
Affiliation(s)
- Gwangwoong Go
- Department of Animal Science, Texas A&M University, College Station, TX 2471, USA
| | | | | | | | | | | |
Collapse
|
49
|
Visioli F, Giordano E, Nicod NM, Dávalos A. Molecular targets of omega 3 and conjugated linoleic Fatty acids - "micromanaging" cellular response. Front Physiol 2012; 3:42. [PMID: 22393325 PMCID: PMC3289952 DOI: 10.3389/fphys.2012.00042] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 02/13/2012] [Indexed: 12/11/2022] Open
Abstract
Essential fatty acids cannot be synthesized de novo by mammals and need to be ingested either with the diet or through the use of supplements/functional foods to ameliorate cardiovascular prognosis. This review focus on the molecular targets of omega 3 fatty acids and conjugated linoleic acid, as paradigmatic molecules that can be exploited both as nutrients and as pharmacological agents, especially as related to cardioprotection. In addition, we indicate novel molecular targets, namely microRNAs that might contribute to the observed biological activities of such essential fatty acids.
Collapse
|
50
|
Wu SJ, Lee SJ, Su CH, Lin DL, Wang SS, Ng LT. Bioactive constituents and anti-proliferative properties of supercritical carbon dioxide Salvia miltiorrhiza extract in 3T3-L1 adipocytes. Process Biochem 2012. [DOI: 10.1016/j.procbio.2011.10.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|