1
|
Magwenyane AM, Kumalo HM. Computational Approaches for PPARγ Inhibitor Development: Recent Advances and Perspectives. ChemistryOpen 2025:e2500087. [PMID: 40326962 DOI: 10.1002/open.202500087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/26/2025] [Indexed: 05/07/2025] Open
Abstract
The development of peroxisome proliferator-activated receptor gamma (PPARγ) inhibitors has attracted significant interest for treating metabolic disorders, cancer, and inflammatory diseases. This review highlights the crucial role of computational modelling in advancing PPARγ inhibitor development, emphasizing how these techniques streamline the identification, optimization, and evaluation of new drug candidates. Key methods include molecular docking, QSAR, and molecular dynamics simulations, which enhance the efficiency and accuracy of inhibitor design. Computational modelling has deepened our understanding of PPARγ binding mechanisms and conformational dynamics, allowing researchers to predict and optimize ligand-receptor complex stability. Despite these advancements, challenges remain, such as improving predictions of pharmacokinetic properties (ADME) to evaluate drug-like qualities. In conclusion, computational modelling has significantly enhanced PPARγ inhibitor discovery and development, offering new opportunities to address complex diseases. Continued refinement of these models, combined with experimental validation and emerging technologies, is crucial for overcoming current limitations and achieving successful clinical outcomes.
Collapse
Affiliation(s)
- Ayanda M Magwenyane
- Chemistry Department, Faculty of Applied and Health Sciences, Mangosuthu University of Technology, Durban, 4031, South Africa
| | - Hezekiel M Kumalo
- Drug Research and Innovation Unit, Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, 4000, South Africa
| |
Collapse
|
2
|
Mansoori S, Ho MY, Ng KK, Cheng KK. Branched-chain amino acid metabolism: Pathophysiological mechanism and therapeutic intervention in metabolic diseases. Obes Rev 2025; 26:e13856. [PMID: 39455059 PMCID: PMC11711082 DOI: 10.1111/obr.13856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 09/02/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024]
Abstract
Branched-chain amino acids (BCAAs), including leucine, isoleucine, and valine, are essential for maintaining physiological functions and metabolic homeostasis. However, chronic elevation of BCAAs causes metabolic diseases such as obesity, type 2 diabetes (T2D), and metabolic-associated fatty liver disease (MAFLD). Adipose tissue, skeletal muscle, and the liver are the three major metabolic tissues not only responsible for controlling glucose, lipid, and energy balance but also for maintaining BCAA homeostasis. Under obese and diabetic conditions, different pathogenic factors like pro-inflammatory cytokines, lipotoxicity, and reduction of adiponectin and peroxisome proliferator-activated receptors γ (PPARγ) disrupt BCAA metabolism, leading to excessive accumulation of BCAAs and their downstream metabolites in metabolic tissues and circulation. Mechanistically, BCAAs and/or their downstream metabolites, such as branched-chain ketoacids (BCKAs) and 3-hydroxyisobutyrate (3-HIB), impair insulin signaling, inhibit adipogenesis, induce inflammatory responses, and cause lipotoxicity in the metabolic tissues, resulting in multiple metabolic disorders. In this review, we summarize the latest studies on the metabolic regulation of BCAA homeostasis by the three major metabolic tissues-adipose tissue, skeletal muscle, and liver-and how dysregulated BCAA metabolism affects glucose, lipid, and energy balance in these active metabolic tissues. We also summarize therapeutic approaches to restore normal BCAA metabolism as a treatment for metabolic diseases.
Collapse
Affiliation(s)
- Shama Mansoori
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Melody Yuen‐man Ho
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Kelvin Kwun‐wang Ng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
| | - Kenneth King‐yip Cheng
- Department of Health Technology and InformaticsThe Hong Kong Polytechnic UniversityHong Kong, China
- Hong Kong Polytechnic University Shenzhen Research InstituteShenzhenChina
| |
Collapse
|
3
|
Cai C, Huang Y, Zhang L, Zhang L. Structural Basis of the Dehydratase Module (hDH) of Human Fatty Acid Synthase. Chembiochem 2024; 25:e202400466. [PMID: 38955950 DOI: 10.1002/cbic.202400466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024]
Abstract
The human fatty acid synthase (hFASN) produces fatty acids for cellar membrane construction, energy storage, biomolecule modifications and signal transduction. Abnormal expression and functions of hFASN highly associate with numerous human diseases such as obesity, diabetes, and cancers, and thereby it has been considered as a valuable potential drug target. So far, the structural and catalytic mechanisms of most of the hFASN enzymatic modules have been extensively studied, except the key dehydratase module (hDH). Here we presented the enzymatic characterization and the high-resolution crystal structure of hDH. We demonstrated that the hDH preferentially catalyzes the acyl substrates with short lengths between 4 to 8-carbons, and exhibits much lower enzymatic activity on longer substrates. Subsequent structural study showed that hDH displays a pseudo-dimeric organization with a single L-shaped composite hydrophobic catalytic tunnel as well as an atypical ACP binding site nearby, indicating that hDH achieves distinct substrate recognition and dehydration mechanisms compared to the conventional bacterial fatty acid dehydratases identified. Our findings laid the foundation for understanding the biological and pathogenic functions of hFASN, and may facilitate therapeutical drug development against diseases with abnormal functionality of hFASN.
Collapse
Affiliation(s)
- Chang Cai
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Yuzhou Huang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Lin Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Liang Zhang
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Systems Medicine for Cancer, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
- Department of Chemical Biology, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
4
|
Zhuang Z, Jia W, Wu L, Li Y, Lu Y, Xu M, Bai H, Bi Y, Wang Z, Chen S, Jiang Y, Chang G. Threonine Deficiency Increases Triglyceride Deposition in Primary Duck Hepatocytes by Reducing STAT3 Phosphorylation. Int J Mol Sci 2024; 25:8142. [PMID: 39125712 PMCID: PMC11312044 DOI: 10.3390/ijms25158142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/12/2024] Open
Abstract
Liver lipid metabolism disruption significantly contributes to excessive fat buildup in waterfowl. Research suggests that the supplementation of Threonine (Thr) in the diet can improve liver lipid metabolism disorder, while Thr deficiency can lead to such metabolic disorders in the liver. The mechanisms through which Thr regulates lipid metabolism remain unclear. STAT3 (signal transducer and activator of transcription 3), a crucial transcription factor in the JAK-STAT (Janus kinase-signal transducer and activator of transcription) pathway, participates in various biological processes, including lipid and energy metabolism. This research investigates the potential involvement of STAT3 in the increased lipid storage seen in primary duck hepatocytes as a result of a lack of Thr. Using small interfering RNA and Stattic, a specific STAT3 phosphorylation inhibitor, we explored the impact of STAT3 expression patterns on Thr-regulated lipid synthesis metabolism in hepatocytes. Through transcriptome sequencing, we uncovered pathways related to lipid synthesis and metabolism jointly regulated by Thr and STAT3. The results showed that Thr deficiency increases lipid deposition in primary duck hepatocytes (p < 0.01). The decrease in protein and phosphorylation levels of STAT3 directly caused this deposition (p < 0.01). Transcriptomic analysis revealed that Thr deficiency and STAT3 knockdown jointly altered the mRNA expression levels of pathways related to long-chain fatty acid synthesis and energy metabolism (p < 0.05). Thr deficiency, through mediating STAT3 inactivation, upregulated ELOVL7, PPARG, MMP1, MMP13, and TIMP4 mRNA levels, and downregulated PTGS2 mRNA levels (p < 0.01). In summary, these results suggest that Thr deficiency promotes lipid synthesis, reduces lipid breakdown, and leads to lipid metabolism disorders and triglyceride deposition by downregulating STAT3 activity in primary duck hepatocytes.
Collapse
Affiliation(s)
- Zhong Zhuang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Wenqian Jia
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Lei Wu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yongpeng Li
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yijia Lu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Minghong Xu
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Hao Bai
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China;
| | - Yulin Bi
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Zhixiu Wang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Shihao Chen
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Yong Jiang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| | - Guobin Chang
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (Z.Z.); (W.J.); (L.W.); (Y.L.); (Y.L.); (M.X.); (Y.B.); (Z.W.); (S.C.); (G.C.)
| |
Collapse
|
5
|
Zhao T, Tian T, Yu H, Cao C, Zhang Z, He Z, Ma Z, Cai R, Li F, Pang W. Identification of porcine fast/slow myogenic exosomes and their regulatory effects on lipid accumulation in intramuscular adipocytes. J Anim Sci Biotechnol 2024; 15:73. [PMID: 38824596 PMCID: PMC11144342 DOI: 10.1186/s40104-024-01029-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 04/01/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Pork quality is affected by the type of muscle fibers, which is closely related to meat color, tenderness and juiciness. Exosomes are tiny vesicles with a diameter of approximately 30-150 nm that are secreted by cells and taken up by recipient cells to mediate communication. Exosome-mediated muscle-fat tissue crosstalk is a newly discovered mechanism that may have an important effect on intramuscular fat deposition and with that on meat quality. Various of adipose tissue-derived exosomes have been discovered and identified, but the identification and function of muscle exosomes, especially porcine fast/slow myotube exosomes, remain unclear. Here, we first isolated and identified exosomes secreted from porcine extensor digitorum longus (EDL) and soleus (SOL), which represent fast and slow muscle, respectively, and further explored their effects on lipid accumulation in longissimus dorsi adipocytes. RESULTS Porcine SOL-derived exosomes (SOL-EXO) and EDL-derived exosomes (EDL-EXO) were first identified and their average particle sizes were approximately 84 nm with double-membrane disc- shapes as observed via transmission electron microscopy and scanning electron microscopy. Moreover, the intramuscular fat content of the SOL was greater than that of the EDL at 180 days of age, because SOL intramuscular adipocytes had a stronger lipid-accumulating capacity than those of the EDL. Raman spectral analysis revealed that SOL-EXO protein content was much greater than that of EDL-EXO. Proteomic sequencing identified 72 proteins that were significantly differentially expressed between SOL-EXO and EDL-EXO, 31 of which were downregulated and 41 of which were upregulated in SOL-EXO. CONCLUSIONS Our findings suggest that muscle-fat tissue interactions occur partly via SOL-EXO promoting adipogenic activity of intramuscular adipocytes.
Collapse
Affiliation(s)
- Tiantian Zhao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Tingting Tian
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - He Yu
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Chaoyue Cao
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Ziyi Zhang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zhaozhao He
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Zeqiang Ma
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Rui Cai
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Fengna Li
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, Hunan, China
| | - Weijun Pang
- Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
6
|
He L, Feng X, Hu C, Liu S, Sheng H, Cai B, Ma Y. HOXA9 gene inhibits proliferation and differentiation and promotes apoptosis of bovine preadipocytes. BMC Genomics 2024; 25:358. [PMID: 38605318 PMCID: PMC11007997 DOI: 10.1186/s12864-024-10231-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 03/15/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Hox gene family is an important transcription factor that regulates cell process, and plays a role in the process of adipocytes differentiation and fat deposition. Previous transcriptome sequencing studies have indicated that the Homeobox A9 gene (HOXA9) is a candidate gene for regulating the process of bovine lipid metabolism, but the function and specific mechanism of action remain unclear. Therefore, this study aims to explore the role of HOXA9 in the proliferation, differentiation and apoptosis of bovine preadipocytes through gain-of-function and lose-of-function. RESULT It found HOXA9 highly expressed in bovine adipose tissue, and its expression level changed significantly during adipocytes differentiation process. It gave a hint that HOXA9 may be involved in the process of bovine lipid metabolism. The results of HOXA9 gain-of-function experiments indicated that HOXA9 appeared to act as a negative regulator not only in the differentiation but also in the proliferation of bovine preadipocytes, which is mainly reflected that overexpression of HOXA9 down-regulate the mRNA and protein expression level of PPARγ, CEBPα and FABP4 (P < 0.05). The mRNA expression level of CDK1, CDK2, PCNA, CCNA2, CCNB1, CCND1 and CCNE2, as well as the protein expression of CDK2 also significantly decreased. The decrease of lipid droplets content was the main characteristic of the phenotype (P < 0.01), which further supported the evidence that HOXA9 was a negative regulator of preadipocytes differentiation. The decrease of cell proliferation rate and EdU positive rate, as well as the limitation of transition of preadipocytes from G0/G1 phase to S phase also provided evidence for the inhibition of proliferation. Apart from this above, we noted an interesting phenomenon that overexpression of HOXA9 showed in a significant upregulation of both mRNA and protein level of apoptosis markers, accompanied by a significant increase in cell apoptosis rate. These data led us not to refute the fact that HOXA9 played an active regulatory role in apoptosis. HOXA9 loss-of-function experiments, however, yielded the opposite results. Considering that HOXA9 acts as a transcription factor, we predicted its target genes. Dual luciferase reporter assay system indicated that overexpression of HOXA9 inhibits activity of PCNA promoter. CONCLUSION Taken together, we demonstrated for the first time that HOXA9 played a role as a negative regulatory factor in the differentiation and proliferation of preadipocytes, but played a positive regulatory role in apoptosis, and it may play a regulatory role by targeting PCNA. This study provides basic data for further exploring the regulatory network of intramuscular fat deposition in bovine.
Collapse
Affiliation(s)
- Lixia He
- College of Animal Science and Technology, Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, 750021, Yinchuan, China
| | - Xue Feng
- College of Animal Science and Technology, Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, 750021, Yinchuan, China
| | - Chunli Hu
- College of Animal Science and Technology, Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, 750021, Yinchuan, China
| | - Shuang Liu
- College of Animal Science and Technology, Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, 750021, Yinchuan, China
| | - Hui Sheng
- College of Animal Science and Technology, Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, 750021, Yinchuan, China
| | - Bei Cai
- College of Animal Science and Technology, Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, 750021, Yinchuan, China
| | - Yun Ma
- College of Animal Science and Technology, Key Laboratory of Ruminant Molecular and Cellular Breeding of Ningxia Hui Autonomous Region, Ningxia University, 750021, Yinchuan, China.
| |
Collapse
|
7
|
Zhong Y, Wang Y, Li X, Qin H, Yan S, Rao C, Fan D, Liu D, Deng F, Miao Y, Yang L, Huang K. PRMT4 Facilitates White Adipose Tissue Browning and Thermogenesis by Methylating PPARγ. Diabetes 2023; 72:1095-1111. [PMID: 37216643 PMCID: PMC10382653 DOI: 10.2337/db22-1016] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 05/10/2023] [Indexed: 05/24/2023]
Abstract
Obesity is a global health threat, and the induction of white adipose tissue (WAT) browning presents a promising therapeutic method for it. Recent publications revealed the essential role of protein arginine methyltransferase 4 (PRMT4) in lipid metabolism and adipogenesis, but its involvement in WAT browning has not been investigated. Our initial studies found that the expression of PRMT4 in adipocytes was upregulated in cold-induced WAT browning but downregulated in obesity. Besides, PRMT4 overexpression in inguinal adipose tissue accelerated WAT browning and thermogenesis to protect against high-fat diet-induced obesity and metabolic disruptions. Mechanistically, our work demonstrated that PRMT4 methylated peroxisome proliferator-activated receptor-γ (PPARγ) on Arg240 to enhance its interaction with the coactivator PR domain-containing protein 16 (PRDM16), leading to the increased expression of thermogenic genes. Taken together, our results uncover the essential role of the PRMT4/PPARγ/PRDM16 axis in the pathogenesis of WAT browning. ARTICLE HIGHLIGHTS Protein arginine methyltransferase 4 (PRMT4) expression was upregulated during cold exposure and negatively correlated with body mass of mice and humans. PRMT4 overexpression in inguinal white adipose tissue of mice improved high-fat diet-induced obesity and associated metabolic impairment due to enhanced heat production. PRMT4 methylated peroxisome proliferator-activated receptor-γ on Arg240 and facilitated the binding of the coactivator PR domain-containing protein 16 to initiate adipose tissue browning and thermogenesis. PRMT4-dependent methylation of peroxisome proliferator-activated receptor-γ on Arg240 is important in the process of inguinal white adipose tissue browning.
Collapse
Affiliation(s)
- Yi Zhong
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yilong Wang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaoguang Li
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haojie Qin
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Yan
- Heart Center and Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Caijun Rao
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Di Fan
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Duqiu Liu
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Liyuan Cardiovascular Center, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Deng
- Department of Urology, The Second Xiangya Hospital, Central South University, Hunan, China
| | - Yanli Miao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ling Yang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology,Wuhan, China
| |
Collapse
|
8
|
Li Q, Wang O, Ji B, Zhao L, Zhao L. Alcohol, White Adipose Tissue, and Brown Adipose Tissue: Mechanistic Links to Lipogenesis and Lipolysis. Nutrients 2023; 15:2953. [PMID: 37447280 PMCID: PMC10346806 DOI: 10.3390/nu15132953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
According to data from the World Health Organization, there were about 3 million deaths caused by alcohol consumption worldwide in 2016, of which about 50% were related to liver disease. Alcohol consumption interfering with the normal function of adipocytes has an important impact on the pathogenesis of alcoholic liver disease. There has been increasing recognition of the crucial role of adipose tissue in regulating systemic metabolism, far beyond that of an inert energy storage organ in recent years. The endocrine function of adipose tissue is widely recognized, and the significance of the proteins it produces and releases is still being investigated. Alcohol consumption may affect white adipose tissue (WAT) and brown adipose tissue (BAT), which interact with surrounding tissues such as the liver and intestines. This review briefly introduces the basic concept and classification of adipose tissue and summarizes the mechanism of alcohol affecting lipolysis and lipogenesis in WAT and BAT. The adipose tissue-liver axis is crucial in maintaining lipid homeostasis within the body. Therefore, this review also demonstrates the effects of alcohol consumption on the adipose tissue-liver axis to explore the role of alcohol consumption in the crosstalk between adipose tissue and the liver.
Collapse
Affiliation(s)
- Qing Li
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China;
| | - Ou Wang
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China;
| | - Baoping Ji
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science & Nutritional Engineering, China Agricultural University, Beijing 100083, China;
| | - Liang Zhao
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology and Business University, Beijing 100048, China;
| | - Lei Zhao
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
9
|
Jiang S, Meng X, Gu H, Sun J, Chen S, Chen Z, Liu D, Liang X. STAU1 promotes adipogenesis by regulating the alternative splicing of Pparγ2 mRNA. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159293. [PMID: 36871938 DOI: 10.1016/j.bbalip.2023.159293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 03/07/2023]
Abstract
During adipocyte differentiation, specific genes such as peroxisome proliferator-activated receptor γ (PPARγ) are transcribed and post-transcriptional pre-mRNA is processed into mature mRNA. Since Pparγ2 pre-mRNAs contain putative binding sites for STAUFEN1 (STAU1), which can affect the alternative splicing of pre-mRNA, we hypothesized that STAU1 might regulate the alternative splicing of Pparγ2 pre-mRNA. In this study, we found that STAU1 affects the differentiation of 3 T3-L1 pre-adipocytes. Through RNA-seq analysis, we confirmed that STAU1 can regulate alternative splicing events during adipocyte differentiation, mainly through exon skipping, which suggests that STAU1 is mainly involved in exon splicing. In addition, gene annotation and cluster analysis revealed that the genes affected by alternative splicing were enriched in lipid metabolism pathways. We further demonstrated that STAU1 can regulate the alternative splicing of Pparγ2 pre-mRNA and affect the splicing of exon E1 through RNA immuno-precipitation, photoactivatable ribonucleotide enhanced crosslinking and immunoprecipitation, and sucrose density gradient centrifugation assays. Finally, we confirmed that STAU1 can regulate the alternative splicing of Pparγ2 pre-mRNA in stromal vascular fraction cells. In summary, this study improves our understanding of the function of STAU1 in adipocyte differentiation and the regulatory network of adipocyte differentiation-related gene expression.
Collapse
Affiliation(s)
- Shuo Jiang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Xuanyu Meng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Functional Center, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Hao Gu
- Department of Laparoscopic Surgery, First Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang 830002, China
| | - Jialei Sun
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Siyuan Chen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Zhe Chen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China
| | - Dihui Liu
- Pathology Center, Xinjiang Medical University Affiliated Tumor Hospital, Urumqi, Xinjiang 830002, China
| | - Xiaodi Liang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, 830017, China.
| |
Collapse
|
10
|
Zhu J, Wilding JP, Hu J. Adipocytes in obesity: A perfect reservoir for SARS-CoV-2? Med Hypotheses 2023; 171:111020. [PMID: 36742015 PMCID: PMC9889082 DOI: 10.1016/j.mehy.2023.111020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/17/2022] [Accepted: 12/30/2022] [Indexed: 01/28/2023]
Abstract
Research evidence suggests that adipocytes in obesity might facilitate SARS-CoV-2 replication, for it was only found in adipose tissue of individuals with overweight or obesity but not lean individuals who died from COVID-19. As lipid metabolism is key to adipocyte function, and viruses are capable of exploiting and manipulating lipid metabolism of host cells for their own benefit of infection, we hypothesize that adipocytes could not only impair host immune defense against viral infection, but also facilitate SARS-CoV-2 entry, replication and assembly as a reservoir to boost the viral infection in obesity. The latter of which could mainly be mediated by SARS-CoV-2 hijacking the abnormal lipid metabolism in the adipocytes. If these were to be confirmed, an approach to combat COVID-19 in people with obesity by taking advantage of the abnormal lipid metabolism in adipocytes might be considered, as well as modifying lipid metabolism of other host cells as a potential adjunctive treatment for COVID-19.
Collapse
Key Words
- ACE2, angiotensin-converting enzyme 2
- ATP, adenosine triphosphate
- Adipocyte
- COVID-19, coronavirus disease 2019
- ER, endoplasmic reticulum
- ERGIC, ER-to-Golgi intermediate compartment
- FFAs, free fatty acids
- LDs, lipid droplets
- Lipid metabolism
- Obesity
- S protein, spike protein
- SARS-CoV-2, severe acute respiratory syndrome coronavirus 2
- Severe acute respiratory syndrome coronavirus 2
- TAGs, triacylglycerols
Collapse
Affiliation(s)
- JingJing Zhu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People’s Republic of China,Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, United Kingdom
| | - John P.H. Wilding
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, United Kingdom
| | - Ji Hu
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People’s Republic of China,Corresponding author
| |
Collapse
|
11
|
Kant R, Chandra L, Verma V, Nain P, Bello D, Patel S, Ala S, Chandra R, Antony MA. Gut microbiota interactions with anti-diabetic medications and pathogenesis of type 2 diabetes mellitus. World J Methodol 2022; 12:246-257. [PMID: 36159100 PMCID: PMC9350729 DOI: 10.5662/wjm.v12.i4.246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/03/2022] [Accepted: 06/18/2022] [Indexed: 02/06/2023] Open
Abstract
Microorganisms including bacteria, viruses, protozoa, and fungi living in the gastrointestinal tract are collectively known as the gut microbiota. Dysbiosis is the imbalance in microbial composition on or inside the body relative to healthy state. Altered Firmicutes to Bacteroidetes ratio and decreased abundance of Akkermansia muciniphila are the predominant gut dysbiosis associated with the pathogenesis of type 2 diabetes mellitus (T2DM) and metabolic syndrome. Pathophysiological mechanisms linking gut dysbiosis, and metabolic diseases and their complications include altered metabolism of short-chain fatty acids and bile acids, interaction with gut hormones, increased gut microbial metabolite trimethylamine-N-oxide, bacterial translocation/Leaky gut syndrome, and endotoxin production such as lipopolysaccharides. The association between the gut microbiota and glycemic agents, however, is much less understood and is the growing focus of research and conversation. Recent studies suggest that the gut microbiota and anti-diabetic medications are interdependent on each other, meaning that while anti-diabetic medications alter the gut microbiota, the gut microbiota also alters the efficacy of anti-diabetic medications. With increasing evidence regarding the significance of gut microbiota, it is imperative to review the role of gut microbiota in the pathogenesis of T2DM. This review also discusses the interaction between gut microbiota and the various medications used in the treatment of T2DM.
Collapse
Affiliation(s)
- Ravi Kant
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, SC 29621, United States
- Department of Endocrinology, Diabetes and Metabolism, AnMed Health, Anderson, SC 29621, United States
| | - Lakshya Chandra
- Department of Internal Medicine, St Francis Hospital, Greenville, SC 29601, United States
| | - Vipin Verma
- Department of Internal Medicine, Medical University of South Carolina, Anderson, SC 29621, United States
- Department of Internal Medicine, AnMed Health, Anderson, SC 29621, United States
| | - Priyanshu Nain
- Department of Graduate Medical Education, Maulana Azad Medical College, Delhi 110002, India
| | - Diego Bello
- Department of Surgery, AnMed Health, Anderson, SC 29621, United States
| | - Siddharth Patel
- Department of Internal Medicine, Decatur Morgan Hospital, Decatur, AL 35601, United States
| | - Subash Ala
- Department of Internal Medicine, St Francis Hospital, Greenville, SC 29601, United States
| | - Rashmi Chandra
- Department of Internal Medicine, Medical University of South Carolina, Anderson, SC 29621, United States
| | - Mc Anto Antony
- Department of Endocrinology, Diabetes and Metabolism, Medical University of South Carolina, Anderson, SC 29621, United States
- Department of Endocrinology, Diabetes and Metabolism, AnMed Health, Anderson, SC 29621, United States
| |
Collapse
|
12
|
Kant R, Chandra L, Verma V, Nain P, Bello D, Patel S, Ala S, Chandra R, Antony MA. Gut microbiota interactions with anti-diabetic medications and pathogenesis of type 2 diabetes mellitus. World J Methodol 2022; 12:246-257. [DOI: https:/doi.org/10.5662/wjm.v12.i4.246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/11/2025] Open
|
13
|
Protective role of activating PPARγ in advanced glycation end products-induced impairment of coronary artery vasodilation via inhibiting p38 phosphorylation and reactive oxygen species production. Biomed Pharmacother 2022; 147:112641. [DOI: 10.1016/j.biopha.2022.112641] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 12/23/2022] Open
|
14
|
Nettore IC, Franchini F, Palatucci G, Macchia PE, Ungaro P. Epigenetic Mechanisms of Endocrine-Disrupting Chemicals in Obesity. Biomedicines 2021; 9:biomedicines9111716. [PMID: 34829943 PMCID: PMC8615468 DOI: 10.3390/biomedicines9111716] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
The incidence of obesity has dramatically increased over the last decades. Recently, there has been a growing interest in the possible association between the pandemics of obesity and some endocrine-disrupting chemicals (EDCs), termed “obesogens”. These are a heterogeneous group of exogenous compounds that can interfere in the endocrine regulation of energy metabolism and adipose tissue structure. Oral intake, inhalation, and dermal absorption represent the major sources of human exposure to these EDCs. Recently, epigenetic changes such as the methylation of cytosine residues on DNA, post-translational modification of histones, and microRNA expression have been considered to act as an intermediary between deleterious effects of EDCs and obesity development in susceptible individuals. Specifically, EDCs exposure during early-life development can detrimentally affect individuals via inducing epigenetic modifications that can permanently change the epigenome in the germline, enabling changes to be transmitted to the next generations and predisposing them to a multitude of diseases. The purpose of this review is to analyze the epigenetic alterations putatively induced by chemical exposures and their ability to interfere with the control of energy metabolism and adipose tissue regulation, resulting in imbalances in the control of body weight, which can lead to obesity.
Collapse
Affiliation(s)
- Immacolata Cristina Nettore
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Fabiana Franchini
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Giuseppe Palatucci
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Paolo Emidio Macchia
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via S. Pansini, 80131 Naples, Italy; (I.C.N.); (F.F.); (G.P.); (P.E.M.)
| | - Paola Ungaro
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale del CNR “G. Salvatore”, Via S. Pansini, 80131 Naples, Italy
- Correspondence: ; Tel.: +39-081-770-4795
| |
Collapse
|
15
|
Xiong L, Pei J, Kalwar Q, Wu X, Yan P, Guo X. Fat deposition in yak during different phenological seasons. Livest Sci 2021. [DOI: 10.1016/j.livsci.2021.104671] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Chaudhuri P, Das M, Lodh I, Goswami R. Role of Metabolic Risk Factors, Family History, and Genetic Polymorphisms (PPARγ and TCF7L2) on Type 2 Diabetes Mellitus Risk in an Asian Indian Population. Public Health Genomics 2021; 24:131-138. [PMID: 33784687 DOI: 10.1159/000514506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 01/12/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Women with family history of diabetes (FHD) are at significantly increased risk of developing gestational diabetes mellitus which may eventually lead to type 2 diabetes mellitus (T2DM) in later life. OBJECTIVE This study investigates the role of FHD on metabolic markers and gene polymorphisms and hence on T2DM susceptibility in nondiabetic pregnant women and the subsequent risks in their newborns. MATERIALS AND METHODS The present study was conducted on 200 healthy (nondiabetic and normotensive) adult Asian Indian women, including 100 with and 100 without FHD, living in and around Kolkata, India. During the gestational period, they were studied twice and followed up till delivery. During delivery, both mothers' venous blood and cord blood were collected to estimate serum CRP, glucose, and lipid profiles of the respective mothers and their newborns. Genotyping of PPARγ and TCF7L2 polymorphisms was done from these blood samples. RESULTS A comparison of the metabolic variables among the subjects with and without FHD revealed significant differences among them. We also found close relationship between mothers and their newborn babies in terms of both PPARγ (rs1801282) C/G and TCF7L2 (rs7903146) C/T polymorphisms. More specifically, genotyping results for mothers with FHD and their newborn babies showed high concordance in inheritance of alleles: (i) for PPARγ via the risk allele G (74.0%) which is carried over to the newborn babies (64.5%) and (ii) for TCF7L2 via the risk allele T (73.0%) which is carried over to the newborn babies (68.5%). CONCLUSION This study leads to the conclusion that Asian Indian women population based in Kolkata, India, are ethnically and genetically predisposed to the risk factors of diabetes through FHD, which is reflected in their gestational phase, and it has a significant implication on their birth outcomes.
Collapse
Affiliation(s)
- Plaban Chaudhuri
- Department of Biotechnology, Heritage Institute of Technology, Kolkata, India
| | - Mithun Das
- Department of Anthropology & Tribal Studies, Sidho-Kanho-Birsha University, Purulia, India
| | | | - Riddhi Goswami
- Department of Biotechnology, Heritage Institute of Technology, Kolkata, India
| |
Collapse
|
17
|
Wei X, Zhu Y, Du J, Ma X, Zhao X, Ma Y, Han S, Ma Y. Analysis of ANGPTL8 promoter activity and screening of related transcription factors in bovine. Gene 2021; 784:145594. [PMID: 33766704 DOI: 10.1016/j.gene.2021.145594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 02/25/2021] [Accepted: 03/16/2021] [Indexed: 11/30/2022]
Abstract
Analysing the molecular regulation mechanism of fat deposition in yellow cattle can provide a theoretical basis for the breeding of excellent beef cattle. ANGPTL8 (angiopoietin-like protein 8) promotes the formation of lipid droplets during adipocyte differentiation. To explore the promoter active region of ANGPTL8 and predict potential transcription factors, we further provide a theoretical basis for the functional analysis and regulatory mechanism of ANGPTL8 in adipogenesis. The promoter region of bovine ANGPTL8 was cloned by overlap extension PCR. Online software was used to predict potential transcription factor binding sites, and it identified PPARγ, SREBP1, C/EBPα, and Znf423 transcription factor binding sites in ANGPTL8 promoter region. A luciferase reporter gene vector which contained different deletion fragments of the ANGPTL8 promoter was constructed. Then, the vectors were cotransfected into 293 T cells with the internal control plasmid pRL-TK by cationic liposomes, and the relative fluorescence intensity was detected by a microplate reader. The results of the luciferase activity analysis showed that the core promoter area of ANGPTL8 was in the -885/-227 bp region of the 5' flanking sequence, while just two SREBP1 binding sites occurred in this area. When SREBP1 was knocked down by siRNA, the expression level of ANGPTL8 was reduced, and we speculated that SREBP1 may be an important transcription factor regulating ANGPTL8 transcription.
Collapse
Affiliation(s)
- Xuefeng Wei
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China
| | - Yunchang Zhu
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China
| | - Jie Du
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China
| | - Xiaojie Ma
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China
| | - Xue Zhao
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China
| | - Yaoyao Ma
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China
| | - Shuang Han
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China
| | - Yun Ma
- College of Life Sciences, Xinyang Normal University, Xinyang, Henan 464000, China; School of Agriculture, Ningxia University, Yinchuan, Ningxia 750021, China.
| |
Collapse
|
18
|
A Comparison of Gene Expression Changes in the Blood of Individuals Consuming Diets Supplemented with Olives, Nuts or Long-Chain Omega-3 Fatty Acids. Nutrients 2020; 12:nu12123765. [PMID: 33302351 PMCID: PMC7762614 DOI: 10.3390/nu12123765] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/13/2022] Open
Abstract
Background: The Mediterranean diet, which is rich in olive oil, nuts, and fish, is considered healthy and may reduce the risk of chronic diseases. Methods: Here, we compared the transcriptome from the blood of subjects with diets supplemented with olives, nuts, or long-chain omega-3 fatty acids and identified the genes differentially expressed. The dietary genes obtained were subjected to network analysis to determine the main pathways, as well as the transcription factors and microRNA interaction networks to elucidate their regulation. Finally, a gene-associated disease interaction network was performed. Results: We identified several genes whose expression is altered after the intake of components of the Mediterranean diets compared to controls. These genes were associated with infection and inflammation. Transcription factors and miRNAs were identified as potential regulators of the dietary genes. Interestingly, caspase 1 and sialophorin are differentially expressed in the opposite direction after the intake of supplements compared to Alzheimer’s disease patients. In addition, ten transcription factors were identified that regulated gene expression in supplemented diets, mild cognitive impairment, and Alzheimer’s disease. Conclusions: We identified genes whose expression is altered after the intake of the supplements as well as the transcription factors and miRNAs involved in their regulation. These genes are associated with schizophrenia, neoplasms, and rheumatic arthritis, suggesting that the Mediterranean diet may be beneficial in reducing these diseases. In addition, the results suggest that the Mediterranean diet may also be beneficial in reducing the risk of dementia.
Collapse
|
19
|
Ginsenosides for the treatment of metabolic syndrome and cardiovascular diseases: Pharmacology and mechanisms. Biomed Pharmacother 2020; 132:110915. [DOI: 10.1016/j.biopha.2020.110915] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/05/2020] [Accepted: 10/17/2020] [Indexed: 12/16/2022] Open
|
20
|
Peroxisome Proliferator-Activated Receptors as Molecular Links between Caloric Restriction and Circadian Rhythm. Nutrients 2020; 12:nu12113476. [PMID: 33198317 PMCID: PMC7696073 DOI: 10.3390/nu12113476] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
The circadian rhythm plays a chief role in the adaptation of all bodily processes to internal and environmental changes on the daily basis. Next to light/dark phases, feeding patterns constitute the most essential element entraining daily oscillations, and therefore, timely and appropriate restrictive diets have a great capacity to restore the circadian rhythm. One of the restrictive nutritional approaches, caloric restriction (CR) achieves stunning results in extending health span and life span via coordinated changes in multiple biological functions from the molecular, cellular, to the whole-body levels. The main molecular pathways affected by CR include mTOR, insulin signaling, AMPK, and sirtuins. Members of the family of nuclear receptors, the three peroxisome proliferator-activated receptors (PPARs), PPARα, PPARβ/δ, and PPARγ take part in the modulation of these pathways. In this non-systematic review, we describe the molecular interconnection between circadian rhythm, CR-associated pathways, and PPARs. Further, we identify a link between circadian rhythm and the outcomes of CR on the whole-body level including oxidative stress, inflammation, and aging. Since PPARs contribute to many changes triggered by CR, we discuss the potential involvement of PPARs in bridging CR and circadian rhythm.
Collapse
|
21
|
Han X, Meng F, Cao X, Du X, Bu G, Kong F, Huang A, Zeng X. FSH promotes fat accumulation by activating PPARγ signaling in surgically castrated, but not immunocastrated, male pigs. Theriogenology 2020; 160:10-17. [PMID: 33166850 DOI: 10.1016/j.theriogenology.2020.10.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Follicle-stimulating hormone (FSH) was recently implicated as a novel regulator of fat accumulation. Surgical castration causes high FSH concentrations and increases fat accumulation, whereas immunocastration results in low FSH concentrations and less fat in immunocastrated boars versus barrows. However, detailed information regarding the role of FSH in regulation of fat accumulation in male pigs is unclear. First, expression of FSH receptor was confirmed (real-time quantitative PCR) in subcutaneous and visceral adipose tissues (SAT and VAT, respectively) of boars. Then, surgical castration (high FSH model) was compared to immunocastration (low FSH model) to investigate potential roles of FSH in adipogenesis and fat accumulation. High FSH concentrations after surgical castration activated PPARγ signaling by upregulating expression of CREB (P < 0.05), and then recruited an array of PPARγ target adipogenic genes, including transcription factor (C/EBPα), long-chain fatty acid uptake (LPL), fatty acid de novo synthesis (FASN, ACACA) and lipid droplet formation (PLIN1) in both SAT and VAT, promoting fat accumulation in barrows. In contrast, much lower serum FSH concentrations in immunocastrates attenuated (P < 0.05) expressions of PPARγ and PPARγ target genes in both SAT and VAT, resulting in less fat accumulation in immunocastrated boars versus barrows. We concluded that the substantially elevated FSH concentrations in barrows promoted fat accumulation by activating the PPARγ signaling pathway in adipose tissues, whereas immunocastrates accumulated less fat due to low FSH.
Collapse
Affiliation(s)
- Xingfa Han
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Fengyan Meng
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Xiaohan Cao
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Xiaogang Du
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Guixian Bu
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Fanli Kong
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Anqi Huang
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China
| | - Xianyin Zeng
- Isotope Research Lab, Sichuan Agricultural University, Ya'an 625014, People's Republic of China.
| |
Collapse
|
22
|
Dim Light at Night Disturbs Molecular Pathways of Lipid Metabolism. Int J Mol Sci 2020; 21:ijms21186919. [PMID: 32967195 PMCID: PMC7555372 DOI: 10.3390/ijms21186919] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/13/2022] Open
Abstract
Dim light at night (dLAN) is associated with metabolic risk but the specific effects on lipid metabolism have only been evaluated to a limited extent. Therefore, to explore whether dLAN can compromise lipid metabolic homeostasis in healthy individuals, we exposed Wistar rats to dLAN (~2 lx) for 2 and 5 weeks and analyzed the main lipogenic pathways in the liver and epididymal fat pad, including the control mechanisms at the hormonal and molecular level. We found that dLAN promoted hepatic triacylglycerol accumulation, upregulated hepatic genes involved in de novo synthesis of fatty acids, and elevated glucose and fatty acid uptake. These observations were paralleled with suppressed fatty acid synthesis in the adipose tissue and altered plasma adipokine levels, indicating disturbed adipocyte metabolic function with a potential negative impact on liver metabolism. Moreover, dLAN-exposed rats displayed an elevated expression of two peroxisome proliferator-activated receptor family members (Pparα and Pparγ) in the liver and adipose tissue, suggesting the deregulation of important metabolic transcription factors. Together, our results demonstrate that an impaired balance of lipid biosynthetic pathways caused by dLAN can increase lipid storage in the liver, thereby accounting for a potential linking mechanism between dLAN and metabolic diseases.
Collapse
|
23
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
24
|
Fougerat A, Montagner A, Loiseau N, Guillou H, Wahli W. Peroxisome Proliferator-Activated Receptors and Their Novel Ligands as Candidates for the Treatment of Non-Alcoholic Fatty Liver Disease. Cells 2020; 9:E1638. [PMID: 32650421 PMCID: PMC7408116 DOI: 10.3390/cells9071638] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/26/2020] [Accepted: 07/04/2020] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a major health issue worldwide, frequently associated with obesity and type 2 diabetes. Steatosis is the initial stage of the disease, which is characterized by lipid accumulation in hepatocytes, which can progress to non-alcoholic steatohepatitis (NASH) with inflammation and various levels of fibrosis that further increase the risk of developing cirrhosis and hepatocellular carcinoma. The pathogenesis of NAFLD is influenced by interactions between genetic and environmental factors and involves several biological processes in multiple organs. No effective therapy is currently available for the treatment of NAFLD. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors that regulate many functions that are disturbed in NAFLD, including glucose and lipid metabolism, as well as inflammation. Thus, they represent relevant clinical targets for NAFLD. In this review, we describe the determinants and mechanisms underlying the pathogenesis of NAFLD, its progression and complications, as well as the current therapeutic strategies that are employed. We also focus on the complementary and distinct roles of PPAR isotypes in many biological processes and on the effects of first-generation PPAR agonists. Finally, we review novel and safe PPAR agonists with improved efficacy and their potential use in the treatment of NAFLD.
Collapse
Affiliation(s)
- Anne Fougerat
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Alexandra Montagner
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Institut National de la Santé et de la Recherche Médicale (Inserm), Institute of Metabolic and Cardiovascular Diseases, UMR1048 Toulouse, France
- Institute of Metabolic and Cardiovascular Diseases, University of Toulouse, UMR1048 Toulouse, France
| | - Nicolas Loiseau
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Hervé Guillou
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
| | - Walter Wahli
- Institut National de la Recherche Agronomique (INRAE), ToxAlim, UMR1331 Toulouse, France; (A.M.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
25
|
Yan K, Wang X, Zhu H, Pan H, Wang L, Yang H, Liu M, Jin M, Zang B, Gong F. Safflower yellow improves insulin sensitivity in high-fat diet-induced obese mice by promoting peroxisome proliferator-activated receptor-γ2 expression in subcutaneous adipose tissue. J Diabetes Investig 2020; 11:1457-1469. [PMID: 32356607 PMCID: PMC7610129 DOI: 10.1111/jdi.13285] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/24/2020] [Accepted: 04/26/2020] [Indexed: 12/13/2022] Open
Abstract
Aims/Introduction Safflower yellow (SY) and its main component, hydroxysafflor yellow A, have been demonstrated to show anti‐obesity effects. Peroxisome proliferator‐activated receptor‐γ2 (PPARγ2) is a critical transcription factor in adipose tissue metabolism. The aim of the present study was to explore the effects of SY in high‐fat diet‐induced obese mice, and further investigate the mechanism involving PPARγ2. Methods High‐fat diet‐induced obese mice were given 120 mg/kg/day SY for 8 weeks. Glucose and insulin tolerance tests were carried out. Fat mass and serum levels of glucose and insulin were measured. The expression of insulin signaling pathway‐related genes and PPARγ2 in the adipose tissue was measured. In vitro, the effects of SY (0–500 mg/L) and hydroxysafflor yellow A (0–100 mg/L) on PPARγ2 promoter activities and PPARγ2 messenger ribonucleic acid (mRNA) levels in 3T3‐L1 preadipocytes or adipocytes were also detected. Results Safflower yellow reduced fat mass, decreased glucose levels and improved insulin sensitivity in obese mice. SY also increased the mRNA levels of insulin signaling pathway‐related genes, and increased PPARγ2 mRNA levels by 39.1% in subcutaneous adipose tissue (P < 0.05). In vitro, SY and hydroxysafflor yellow A significantly enhanced PPARγ2 promoter activities by 1.3–2.1‐fold, and increased PPARγ2 mRNA levels by 1.2–1.6‐fold in 3T3‐L1 preadipocytes or adipocytes (P < 0.05). Conclusions SY could reduce fat mass, decrease glucose levels and improve insulin sensitivity in high‐fat diet‐induced obese mice. The probable mechanism is to increase PPARγ2 expression by stimulating PPARγ2 promoter activities, further increasing the expression of insulin signaling pathway‐related genes in subcutaneous adipose tissue.
Collapse
Affiliation(s)
- Kemin Yan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiangqing Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Meijuan Liu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Ming Jin
- Department of Pharmacology, China-Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Baoxia Zang
- Department of Pharmacology, China-Beijing Institute of Heart, Lung and Blood Vessel Disease, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Agrimi J, Baroni C, Anakor E, Lionetti V. Perioperative Heart-Brain Axis Protection in Obese Surgical Patients: The Nutrigenomic Approach. Curr Med Chem 2020; 27:258-281. [PMID: 30324875 DOI: 10.2174/0929867325666181015145225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 08/01/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
The number of obese patients undergoing cardiac and noncardiac surgery is rapidly increasing because they are more prone to concomitant diseases, such as diabetes, thrombosis, sleep-disordered breathing, cardiovascular and cerebrovascular disorders. Even if guidelines are already available to manage anesthesia and surgery of obese patients, the assessment of the perioperative morbidity and mortality from heart and brain disorders in morbidly obese surgical patients will be challenging in the next years. The present review will recapitulate the new mechanisms underlying the Heart-brain Axis (HBA) vulnerability during the perioperative period in healthy and morbidly obese patients. Finally, we will describe the nutrigenomics approach, an emerging noninvasive dietary tool, to maintain a healthy body weight and to minimize the HBA propensity to injury in obese individuals undergoing all types of surgery by personalized intake of plant compounds that may regulate the switch from health to disease in an epigenetic manner. Our review provides current insights into the mechanisms underlying HBA response in obese surgical patients and how they are modulated by epigenetically active food constituents.
Collapse
Affiliation(s)
- Jacopo Agrimi
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Carlotta Baroni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Ekene Anakor
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Vincenzo Lionetti
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.,UOS Anesthesiology, Fondazione Toscana G. Monasterio, Pisa, Italy
| |
Collapse
|
27
|
Marbach-Breitrück E, Kutzner L, Rothe M, Gurke R, Schreiber Y, Reddanna P, Schebb NH, Stehling S, Wieler LH, Heydeck D, Kuhn H. Functional Characterization of Knock-In Mice Expressing a 12/15-Lipoxygenating Alox5 Mutant Instead of the 5-Lipoxygenating Wild-Type Enzyme. Antioxid Redox Signal 2020; 32:1-17. [PMID: 31642348 DOI: 10.1089/ars.2019.7751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aims: Most mammalian genomes involve several genes encoding for functionally distinct arachidonate lipoxygenase (ALOX isoforms). Proinflammatory leukotrienes are formed via the ALOX5 pathway, but 12/15-lipoxygenating ALOX isoforms have been implicated in the biosynthesis of pro-resolving mediators. In vitro mutagenesis of the triad determinants abolished the leukotriene synthesizing activity of ALOX5, but the biological consequences of these alterations have not been studied. To fill this gap, we created Alox5 knock-in mice, which express the 12/15-lipoxygenating Phe359Trp + Ala424Ile + Asn425Met Alox5 triple mutant and characterized its phenotypic alterations. Results: The mouse Alox5 triple mutant functions as arachidonic acid 15-lipoxygenating enzyme, which also forms 12S-hydroxy and 8S-hydroxy arachidonic acid. In contrast to the wild-type enzyme, the triple mutant effectively oxygenates linoleic acid to 13S-hydroxy linoleic acid (13S-HODE), which functions as activating ligand of the type-2 nuclear receptor peroxisome proliferator-activated receptor gamma (PPARγ). Knock-in mice expressing the mutant enzyme are viable, fertile, and develop normally. The mice cannot synthesize proinflammatory leukotrienes but show significantly attenuated plasma levels of lipolytic endocannabinoids. When aging, the animals gained significantly more body weight, which may be related to the fivefold higher levels of 13-HODE in the adipose tissue. Innovation: These data indicate for the first time that in vivo mutagenesis of the triad determinants of mouse Alox5 abolished the biosynthetic capacity of the enzyme for proinflammatory leukotrienes and altered the catalytic properties of the protein favoring the formation of 13-HODE. Conclusion:In vivo triple mutation of the mouse Alox5 gene impacts the body weight homeostasis of aging mice via augmented formation of the activating PPARγ ligand 13-HODE.
Collapse
Affiliation(s)
- Eugenia Marbach-Breitrück
- Institute of Biochemistry, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Laura Kutzner
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | | | - Robert Gurke
- Pharmazentrum Frankfurt (ZAFES), Institute of Clinical Pharmacology, Goethe University, Frankfurt am Main, Germany.,Branch for Translational Medicine and Pharmacology (TMP), Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt am Main, Germany
| | - Yannick Schreiber
- Branch for Translational Medicine and Pharmacology (TMP), Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Frankfurt am Main, Germany
| | - Pallu Reddanna
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad India
| | - Nils-Helge Schebb
- Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Sabine Stehling
- Institute of Biochemistry, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Lothar H Wieler
- Robert Koch Institute, Berlin, Germany.,Institute of Microbiology and Epizootics, Center of Infection Medicine, Free University of Berlin, Berlin, Germany
| | - Dagmar Heydeck
- Institute of Biochemistry, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Hartmut Kuhn
- Institute of Biochemistry, Charité-University Medicine Berlin, Corporate Member of Free University Berlin, Humboldt-University zu Berlin, Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
28
|
Chmurzynska A, Muzsik A, Krzyżanowska-Jankowska P, Mądry E, Walkowiak J, Bajerska J. PPARG and FTO polymorphism can modulate the outcomes of a central European diet and a Mediterranean diet in centrally obese postmenopausal women. Nutr Res 2019; 69:94-100. [PMID: 31675538 DOI: 10.1016/j.nutres.2019.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/27/2019] [Accepted: 08/06/2019] [Indexed: 01/15/2023]
Abstract
The aim of this study was to test the hypothesis that polymorphism of genes with the biggest effects on body mass (FTO and PPARG) can affect the results of dieting in centrally obese postmenopausal women. A total of 144 volunteers were randomized to a 16-week intervention with two hypocaloric diets: either a Mediterranean diet (MED) moderate in fat (37% total energy as fat) or the Central European diet (CED) moderate in carbohydrates (55% total energy as carbohydrates). The associations between FTO and PPARG polymorphism on the baseline body mass, body composition, blood pressure, lipid and non-lipid parameters, and their changes after the trial were analyzed. None of the examined baseline outcomes differed in the rs9939609 FTO subgroups; abdominal fat was higher in the minor (G) allele carriers of the PPARG rs1801282. After the intervention, in the CED group, the PPARG G allele carriers showed greater reductions in weight (-6.58 ± 0.61 vs -9.58 ± 0.83; P < .01), lean mass (-0.38 ± 0.29 vs -1.79 ± 0.38; P < .05) and high-density lipoprotein (HDL) cholesterol (-0.46 ± 0.77 vs -5.25 ± 1.49; P < .01) than the CC homozygotes, and the TT individuals of the rs9939609 FTO had greater reductions in diastolic blood pressure (-9.03 ± 1.78 vs. -7.58 ± 1.50; P < .05). In the MED group, greater reductions in abdominal fat were observed in the G allele carriers than in the CC homozygotes (-3.31 ± 0.26 vs. -4.23 ± 0.41; P < .05). PPARG and FTO polymorphism may affect the outcomes of the diets aimed at weight reduction in postmenopausal women.
Collapse
Affiliation(s)
- Agata Chmurzynska
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego 31, 60-624 Poznan, Poland
| | - Agata Muzsik
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego 31, 60-624 Poznan, Poland
| | - Patrycja Krzyżanowska-Jankowska
- First Subdepartment of Pediatrics, Department of Pediatric Gastroenterology and Metabolism, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572 Poznan, Poland
| | - Edyta Mądry
- First Subdepartment of Pediatrics, Department of Pediatric Gastroenterology and Metabolism, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572 Poznan, Poland
| | - Jarosław Walkowiak
- First Subdepartment of Pediatrics, Department of Pediatric Gastroenterology and Metabolism, Poznan University of Medical Sciences, Szpitalna 27/33, 60-572 Poznan, Poland
| | - Joanna Bajerska
- Institute of Human Nutrition and Dietetics, Poznan University of Life Sciences, Wojska Polskiego 31, 60-624 Poznan, Poland.
| |
Collapse
|
29
|
ANGPTL8 regulates adipocytes differentiation and adipogenesis in bovine. Gene 2019; 707:93-99. [PMID: 31048067 DOI: 10.1016/j.gene.2019.04.048] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 03/20/2019] [Accepted: 04/17/2019] [Indexed: 12/17/2022]
Abstract
The formation of bovine adipose tissue involves complex developmental and physiological processes that play a vital role in determining the quality of beef; however, the regulatory mechanisms are largely unknown. Angiopoietin-like protein 8 (ANGPTL8) has been reported to be involved in the development of adipose tissue; however, the mechanism of adipogenesis which is regulated by ANGPTL8 has not been revealed in cattle. In this study, RT-qPCR and Oil Red O staining were performed to detect the expression of ANGPTL8 and adipocyte differentiation in bovine. We found that ANGPTL8 had a high expression level in adipose tissue and that the expression pattern was consistent with those of PPARγ, C/EBPα and LPL which are key regulators and transcription factors involved in preadipocyte differentiation and adipogenesis. The overexpression of ANGPTL8 by the adenovirus vector promoted lipid droplet formation in adipocytes. Thus, we speculated that ANGPTL8 could significantly enhance lipid deposition. Moreover, the expression of LPL and SREBP1, key genes inhibiting adipogenesis, was significantly decreased by ANGPTL8 overexpression. These results suggested that ANGPTL8 promotes adipocyte differentiation. In conclusion, we consider that ANGPTL8 regulates adipocyte differentiation and adipogenesis in bovine.
Collapse
|
30
|
Song Y, Li X, Liu Y, Hu Y, Yang R. Arctigenin improves lipid metabolism by regulating AMP-activated protein kinase and downstream signaling pathways. J Cell Biochem 2019; 120:13275-13288. [PMID: 30891825 DOI: 10.1002/jcb.28602] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/16/2022]
Abstract
Although it has been reported that arctigenin (ARG) can reduce the body weight and inhibit adipogenic differentiation by activating AMP-activated protein kinase (AMPK), the exact signals responsible for the ARG-mediated antiobesity mechanism through AMPK are not well understood. In this study, we investigated the potential improvement of AGR on lipid metabolism using a high-fat diet (HFD)-induced hyperlipidemia rats and 3T3-L1 mature adipocytes. The levels of AMPK and its downstream factors were examined by Western blot analysis and real-time fluorescent quantitative polymerase chain reaction. We observed that ARG lowered the HFD-induced body weight and the levels of serum lipid. Moreover, ARG clearly alleviated fat deposition in the liver and reduced epididymal fat accumulation. ARG also suppressed lipogenesis and lipolysis but promoted fatty acid β-oxidation in adipocytes. Most importantly, ARG increased the phosphorylation of AMPK and acetyl-CoA carboxylase (ACC) and upregulated the messenger RNA levels of downstream genes related to fatty acid β-oxidation, such as carnitine palmitoyltransferase 1 and acyl-CoA oxidase 1 but downregulated the expression of peroxisome proliferator-activated receptor γ (PPARγ), sterol regulatory element-binding transcription factor 1 (SREBP1c) and their targets, including lipogenesis-related genes such as CCAAT/enhancer-binding protein α, lipoprotein lipase, adipocyte protein 2, and fatty acid synthase (FAS), as well as lipolysis-related genes such as adipose triglyceride lipase and hormone-sensitive lipase. The activity of FAS was also decreased by ARG. We conclude that AMPK activation is important for the pharmacological effects of ARG. ARG may improve lipid metabolism by regulating the AMPK-ACC and AMPK-PPARγ/SREBP1c signaling pathways.
Collapse
Affiliation(s)
- Yuzhou Song
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiao Li
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yunyun Liu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yingjie Hu
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Ruiyi Yang
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
31
|
Kyriachenko Y, Falalyeyeva T, Korotkyi O, Molochek N, Kobyliak N. Crosstalk between gut microbiota and antidiabetic drug action. World J Diabetes 2019; 10:154-168. [PMID: 30891151 PMCID: PMC6422856 DOI: 10.4239/wjd.v10.i3.154] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 03/10/2019] [Accepted: 03/11/2019] [Indexed: 02/05/2023] Open
Abstract
Type 2 diabetes (T2D) is a disorder characterized by chronic inflated blood glucose levels (hyperglycemia), at first due to insulin resistance and unregulated insulin secretion but with tendency towards global spreading. The gut microbiota is recognized to have an influence on T2D, although surveys have not formed a clear overview to date. Because of the interactions between gut microbiota and host homeostasis, intestinal bacteria are believed to play a large role in various diseases, including metabolic syndrome, obesity and associated disease. In this review, we highlight the animal and human studies which have elucidated the roles of metformin, α-glucosidase inhibitors, glucagon-like peptide-1 agonists, peroxisome proliferator-activated receptors γ agonists, inhibitors of dipeptidyl peptidase-4, sodium/glucose cotransporter inhibitors, and other less studied medications on gut microbiota. This review is dedicated to one of the most widespread diseases, T2D, and the currently used antidiabetic drugs and most promising new findings. In general, the gut microbiota has been shown to have an influence on host metabolism, food consumption, satiety, glucose homoeostasis, and weight gain. Altered intestinal microbiota composition has been noticed in cardiovascular diseases, colon cancer, rheumatoid arthritis, T2D, and obesity. Therefore, the main effect of antidiabetic drugs is on the microbiome composition, basically increasing the short-chain fatty acids-producing bacteria, responsible for losing weight and suppressing inflammation.
Collapse
Affiliation(s)
- Yevheniia Kyriachenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Tetyana Falalyeyeva
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Oleksandr Korotkyi
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Nataliia Molochek
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, Kyiv 01601, Ukraine
| | - Nazarii Kobyliak
- Endocrinology Department, Bogomolets National Medical University, Kyiv 01601, Ukraine
| |
Collapse
|
32
|
Rharass T, Lucas S. High Glucose Level Impairs Human Mature Bone Marrow Adipocyte Function Through Increased ROS Production. Front Endocrinol (Lausanne) 2019; 10:607. [PMID: 31551934 PMCID: PMC6746912 DOI: 10.3389/fendo.2019.00607] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022] Open
Abstract
Bone marrow adipocytes (BMAds) accumulate in aging, menopause, and metabolic diseases such as Type 2 diabetes. These osteoporotic conditions are associated with oxidative stress and hyperglycemia which are both considered as critical factors underlying bone fragility. Glucose excess and reactive oxygen species (ROS) are known to favor adipogenesis over osteoblastogenesis. In this study, we investigated whether high glucose exposure could determine dysfunction of mature BMAds, specifically through ROS production. The effects of low (LG, 5 mM) or high glucose (HG, 25 mM) concentrations were examined using human bone mesenchymal stromal cells (hBMSCs) in the time course of differentiation, and, up to 21 days once adipocytes were mature. HG did not alter the adipocyte differentiation process of hBMSCs. Yet, after 21 days under HG exposure, PPARG, CEBPA, and adiponectin mRNA expressions were decreased. These alterations were also observed following adipogenic inducer withdrawal as well as in adipocytes fully differentiated in LG then cultured in HG for the last 11 days. Without inducers, HG condition also led to decreased leptin mRNA level. Importantly, intracellular and extracellular ROS concentrations measured using Amplex Red were significantly raised by 50% under HG exposure. This rise was observed once adipocytes ended differentiation and was reproduced within the different cell culture settings without any cytotoxicity. Among genes involved in ROS metabolism, the mRNA level of the H2O2 generating enzyme NOX4 was found upregulated in the presence of HG. Following cell separation, mature BMAds were shown to overproduce ROS and to display the gene alterations in contrast to non-lipid-laden cells. Finally, a non-lethal treatment with a pro-oxidant agent under LG condition reduces the mRNA levels of PPARG, adiponectin, and leptin as the HG condition does in the absence of inducers, and amplifies the effect of glucose excess on gene expression. HG concentration drives mature BMAds toward altered expression of the main adipokines and transcriptional factors. These perturbations are associated with a rise in ROS generation likely mediated through enhanced expression of NOX4. Mature BMAds are thus responsive to changes in glucose and ROS concentrations, which is relevant regarding with their phenotype and function in age- or metabolic disease-related osteoporosis.
Collapse
|
33
|
Blanchard PG, Moreira RJ, Castro É, Caron A, Côté M, Andrade ML, Oliveira TE, Ortiz-Silva M, Peixoto AS, Dias FA, Gélinas Y, Guerra-Sá R, Deshaies Y, Festuccia WT. PPARγ is a major regulator of branched-chain amino acid blood levels and catabolism in white and brown adipose tissues. Metabolism 2018; 89:27-38. [PMID: 30316815 DOI: 10.1016/j.metabol.2018.09.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/25/2018] [Accepted: 09/26/2018] [Indexed: 01/14/2023]
Abstract
OBJECTIVE We investigated whether PPARγ modulates adipose tissue BCAA metabolism, and whether this mediates the attenuation of obesity-associated insulin resistance induced by pharmacological PPARγ activation. METHODS Mice with adipocyte deletion of one or two PPARγ copies fed a chow diet and rats fed either chow, or high fat (HF) or HF supplemented with BCAA (HF/BCAA) diets treated with rosiglitazone (30 or 15 mg/kg/day, 14 days) were evaluated for glucose and BCAA homeostasis. RESULTS Adipocyte deletion of one PPARγ copy increased mice serum BCAA and reduced inguinal white (iWAT) and brown (BAT) adipose tissue BCAA incorporation into triacylglycerol, as well as mRNA levels of branched-chain aminotransferase (BCAT)2 and branched-chain α-ketoacid dehydrogenase (BCKDH) complex subunits. Adipocyte deletion of two PPARγ copies induced lipodystrophy, severe glucose intolerance and markedly increased serum BCAA. Rosiglitazone abolished the increase in serum BCAA induced by adipocyte PPARγ deletion. In rats, HF increased serum BCAA, such levels being further increased by BCAA supplementation. Rosiglitazone, independently of diet, lowered serum BCAA and upregulated iWAT and BAT BCAT and BCKDH activities. This was associated with a reduction in mTORC1-dependent inhibitory serine phosphorylation of IRS1 in skeletal muscle and whole-body insulin resistance evaluated by HOMA-IR. CONCLUSIONS PPARγ, through the regulation of both BAT and iWAT BCAA catabolism in lipoeutrophic mice and muscle insulin responsiveness and proteolysis in lipodystrophic mice, is a major determinant of circulating BCAA levels. PPARγ agonism, therefore, may improve whole-body and muscle insulin sensitivity by reducing blood BCAA, alleviating mTORC1-mediated inhibitory IRS1 phosphorylation.
Collapse
Affiliation(s)
- Pierre-Gilles Blanchard
- Department of Medicine, Faculty of Medicine, Quebec Heart & Lung Institute, Laval University, Quebec, Canada
| | - Rafael J Moreira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Érique Castro
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alexandre Caron
- Department of Medicine, Faculty of Medicine, Quebec Heart & Lung Institute, Laval University, Quebec, Canada
| | - Marie Côté
- Department of Medicine, Faculty of Medicine, Quebec Heart & Lung Institute, Laval University, Quebec, Canada
| | - Maynara L Andrade
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Tiago E Oliveira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Milene Ortiz-Silva
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Albert S Peixoto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - France Anne Dias
- Department of Biological Sciences, ICEB, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Yves Gélinas
- Department of Medicine, Faculty of Medicine, Quebec Heart & Lung Institute, Laval University, Quebec, Canada
| | - Renata Guerra-Sá
- Department of Biological Sciences, ICEB, Federal University of Ouro Preto, Ouro Preto, Brazil
| | - Yves Deshaies
- Department of Medicine, Faculty of Medicine, Quebec Heart & Lung Institute, Laval University, Quebec, Canada
| | - William T Festuccia
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
34
|
Liu C, Li L, Guo D, Lv Y, Zheng X, Mo Z, Xie W. Lipoprotein lipase transporter GPIHBP1 and triglyceride-rich lipoprotein metabolism. Clin Chim Acta 2018; 487:33-40. [PMID: 30218660 DOI: 10.1016/j.cca.2018.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 02/05/2023]
Abstract
Increased plasma triglyceride serves as an independent risk factor for cardiovascular disease (CVD). Lipoprotein lipase (LPL), which hydrolyzes circulating triglyceride, plays a crucial role in normal lipid metabolism and energy balance. Hypertriglyceridemia is possibly caused by gene mutations resulting in LPL dysfunction. There are many factors that both positively and negatively interact with LPL thereby impacting TG lipolysis. Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1), a newly identified factor, appears essential for transporting LPL to the luminal side of the blood vessel and offering a platform for TG hydrolysis. Numerous lines of evidence indicate that GPIHBP1 exerts distinct functions and plays diverse roles in human triglyceride-rich lipoprotein (TRL) metabolism. In this review, we discuss the GPIHBP1 gene, protein, its expression and function and subsequently focus on its regulation and provide critical evidence supporting its role in TRL metabolism. Underlying mechanisms of action are highlighted, additional studies discussed and potential therapeutic targets reviewed.
Collapse
Affiliation(s)
- Chuhao Liu
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China; 2016 Class of Excellent Doctor, University of South China, Hengyang 421001, Hunan, China
| | - Liang Li
- Department of Pathophysiology, University of South China, Hengyang 421001, Hunan, China
| | - Dongming Guo
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China
| | - Yuncheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China
| | - XiLong Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Health Sciences Center, 3330 Hospital Dr NW, Calgary T2N 4N1, Alberta, Canada; Key Laboratory of Molecular Targets & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, Guangdong, China
| | - Zhongcheng Mo
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China.
| | - Wei Xie
- Clinical Anatomy & Reproductive Medicine Application Institute, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
35
|
Duszka K, Wahli W. Enteric Microbiota⁻Gut⁻Brain Axis from the Perspective of Nuclear Receptors. Int J Mol Sci 2018; 19:ijms19082210. [PMID: 30060580 PMCID: PMC6121494 DOI: 10.3390/ijms19082210] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptors (NRs) play a key role in regulating virtually all body functions, thus maintaining a healthy operating body with all its complex systems. Recently, gut microbiota emerged as major factor contributing to the health of the whole organism. Enteric bacteria have multiple ways to influence their host and several of them involve communication with the brain. Mounting evidence of cooperation between gut flora and NRs is already available. However, the full potential of the microbiota interconnection with NRs remains to be uncovered. Herewith, we present the current state of knowledge on the multifaceted roles of NRs in the enteric microbiota–gut–brain axis.
Collapse
Affiliation(s)
- Kalina Duszka
- Department of Nutritional Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria.
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological, 11 Mandalay Road, Singapore 308232, Singapore.
- Center for Integrative Genomics, University of Lausanne, Génopode, CH-1015 Lausanne, Switzerland.
| |
Collapse
|
36
|
Ribeiro Filho HV, Bernardi Videira N, Bridi AV, Tittanegro TH, Helena Batista FA, de Carvalho Pereira JG, de Oliveira PSL, Bajgelman MC, Le Maire A, Figueira ACM. Screening for PPAR Non-Agonist Ligands Followed by Characterization of a Hit, AM-879, with Additional No-Adipogenic and cdk5-Mediated Phosphorylation Inhibition Properties. Front Endocrinol (Lausanne) 2018; 9:11. [PMID: 29449830 PMCID: PMC5799700 DOI: 10.3389/fendo.2018.00011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 01/11/2018] [Indexed: 11/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a member of a nuclear receptor superfamily and acts as a ligand-dependent transcription factor, playing key roles in maintenance of adipose tissue and in regulation of glucose and lipid homeostasis. This receptor is the target of thiazolidinediones, a class of antidiabetic drugs, which improve insulin sensitization and regulate glycemia in type 2 diabetes. Despite the beneficial effects of drugs, such as rosiglitazone and pioglitazone, their use is associated with several side effects, including weight gain, heart failure, and liver disease, since these drugs induce full activation of the receptor. By contrast, a promising activation-independent mechanism that involves the inhibition of cyclin-dependent kinase 5 (CDK5)-mediated PPARγ phosphorylation has been related to the insulin-sensitizing effects induced by these drugs. Thus, we aimed to identify novel PPARγ ligands that do not possess agonist properties by conducting a mini-trial with 80 compounds using the sequential steps of thermal shift assay, 8-anilino-1-naphthalenesulfonic acid fluorescence quenching, and a cell-based transactivation assay. We identified two non-agonist PPARγ ligands, AM-879 and P11, and one partial-agonist, R32. Using fluorescence anisotropy, we show that AM-879 does not dissociate the NCOR corepressor in vitro, and it has only a small effect on TRAP coactivator recruitment. In cells, AM-879 could not induce adipocyte differentiation or positively regulate the expression of genes associated with adipogenesis. In addition, AM-879 inhibited CDK5-mediated phosphorylation of PPARγ in vitro. Taken together, these findings supported an interaction between AM-879 and PPARγ; this interaction was identified by the analysis of the crystal structure of the PPARγ:AM-879 complex and evidenced by AM-879's mechanism of action as a putative PPARγ non-agonist with antidiabetic properties. Moreover, we present an optimized assay pipeline capable of detecting ligands that physically bind to PPARγ but do not cause its activation as a new strategy to identify ligands for this nuclear receptor.
Collapse
Affiliation(s)
- Helder Veras Ribeiro Filho
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Post Graduation Program in Biosciences and Technology of Bioactive Products, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Natália Bernardi Videira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Post Graduation Program in Biosciences and Technology of Bioactive Products, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Aline Villanova Bridi
- Post Graduation Program in Biosciences and Technology of Bioactive Products, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | - Thais Helena Tittanegro
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Post Graduation Program in Biosciences and Technology of Bioactive Products, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| | | | - José Geraldo de Carvalho Pereira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Paulo Sérgio Lopes de Oliveira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Marcio Chaim Bajgelman
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Albane Le Maire
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Centre de Biochimie Structurale CNRS, Université de Montpellier, Montpellier, France
| | - Ana Carolina Migliorini Figueira
- Brazilian Biosciences National Laboratory (LNBio), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
- Post Graduation Program in Biosciences and Technology of Bioactive Products, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil
| |
Collapse
|
37
|
Huang Q, Ma C, Chen L, Luo D, Chen R, Liang F. Mechanistic Insights Into the Interaction Between Transcription Factors and Epigenetic Modifications and the Contribution to the Development of Obesity. Front Endocrinol (Lausanne) 2018; 9:370. [PMID: 30034368 PMCID: PMC6043677 DOI: 10.3389/fendo.2018.00370] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022] Open
Abstract
Objective: The development of obesity is inseparable from genetic and epigenetic factors, and transcription factors (TFs) play an essential role in these two mechanisms. This review analyzes the interaction of TFs with epigenetic modifications and the epigenetic mechanisms underlying peroxisome proliferator-activated receptor (PPAR)γ, an important transcription factor, in the development of obesity. Methods: We describe the relationship between TFs and different epigenetic modifications and illustrate the several mechanisms described. Next, we summarize the epigenetic mechanisms of PPARs, an important class of transcription factors involved in obesity, that induce obesity with different triggering factors. Finally, we discuss the mechanisms of epigenetic modification of PPAR-related ligands in lipid metabolism and propose future avenues of research. Results: TFs participate in epigenetic modifications in different forms, causing changes in gene expression. The interactions between the different epigenetic modifications and PPARs are important biological developments that affect fat tissue differentiation, lipogenesis, and lipid metabolism, thereby inducing or inhibiting the development of obesity. We then highlight the need for more research to understand the role of epigenetic modifications and PPARs. Conclusions: Epigenetic mechanisms involved in the regulation of PPARs may be excellent therapeutic targets for obesity treatment. However, there is a need for a deeper understanding of how PPARs and other obesity-related transcription factors interact with epigenetic modifications.
Collapse
Affiliation(s)
- Qi Huang
- Department of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan, China
| | - Chaoyang Ma
- Department of Rehabilitation, The Central Hospital of Wuhan, Tongji Medical College of Huazhong Science and Technology University, Wuhan, China
| | - Li Chen
- Department of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan, China
| | - Dan Luo
- Department of Traditional Chinese Medicine, Huazhong University of Science and Technology Tongji Medical College, Wuhan, China
| | - Rui Chen
- Department of Integrated TCM and Western Medicine, Union Hospital, Tongji Medical College of Huazhong Science and Technology University, Wuhan, China
- *Correspondence: Rui Chen
| | - Fengxia Liang
- Department of Acupuncture and Moxibustion, Hubei University of Chinese Medicine, Wuhan, China
- Fengxia Liang
| |
Collapse
|
38
|
Yang X, Wang Q, Pang ZR, Pan MR, Zhang W. Flavonoid-enriched extract from Hippophae rhamnoides seed reduces high fat diet induced obesity, hypertriglyceridemia, and hepatic triglyceride accumulation in C57BL/6 mice. PHARMACEUTICAL BIOLOGY 2017; 55:1207-1214. [PMID: 28248545 PMCID: PMC6130443 DOI: 10.1080/13880209.2016.1278454] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 09/27/2016] [Accepted: 12/30/2016] [Indexed: 05/14/2023]
Abstract
CONTEXT Flavonoid-enriched extract from Hippophae rhamnoides L. (Elaeagnaceae) seed (FSH) has shown beneficial effects in anti-hypertension and lowering cholesterol level. However, evidence for its efficacy in treating obesity is limited. OBJECTIVE We sought to determine if FSH can reduce body weight and regulate lipid metabolism disorder in high fat diet (HFD)-induced obese mouse model, and to investigate potential molecular targets involved. MATERIALS AND METHODS C57BL/6 mice were fed with HFD for 8 weeks to induce obesity. The modeled mice were divided into four groups and treated with vehicle, rosiglitazone (2 mg/kg), low (100 mg/kg) and high (300 mg/kg) dose of FSH, respectively. Normal control was also used. The treatments were administered orally for 9 weeks. We measured the effect of FSH on regulating body weight, various liver and serum parameters, and molecular targets that are key to lipid metabolism. RESULTS FSH administration at 100 and 300 mg/kg significantly reduced body weight gain by 33.06 and 43.51%, respectively. Additionally, triglyceride concentration in serum and liver were decreased by 15.67 and 49.56%, individually, after FSH (300 mg/kg) treatment. Upon FSH (100 and 300 mg/kg) treatment, PPARα mRNA expression was upregulated in liver (1.24- and 1.42-fold) and in adipose tissue (1.66- and 1.72-fold). Furthermore, FSH downregulated PPARγ protein level both in liver and adipose tissue. Moreover, FSH inhibited macrophage infiltration into adipose tissues, and downregulated TNFα mRNA expression in adipose tissue (38.01-47.70%). CONCLUSION This effect was mediated via regulation of PPARγ and PPARα gene expression, and suppression of adipose tissue inflammation.
Collapse
Affiliation(s)
- Xin Yang
- School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Qian Wang
- Institute of Oncology, Shanghai 9th People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Zeng-run Pang
- School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Meng-ran Pan
- School of Life Sciences, East China Normal University, Shanghai, P.R. China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai, P.R. China
| |
Collapse
|
39
|
PPARγ Modulates Long Chain Fatty Acid Processing in the Intestinal Epithelium. Int J Mol Sci 2017; 18:ijms18122559. [PMID: 29182565 PMCID: PMC5751162 DOI: 10.3390/ijms18122559] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 11/24/2017] [Accepted: 11/27/2017] [Indexed: 12/26/2022] Open
Abstract
Nuclear receptor PPARγ affects lipid metabolism in several tissues, but its role in intestinal lipid metabolism has not been explored. As alterations have been observed in the plasma lipid profile of ad libitum fed intestinal epithelium-specific PPARγ knockout mice (iePPARγKO), we submitted these mice to lipid gavage challenges. Within hours after gavage with long chain unsaturated fatty acid (FA)-rich canola oil, the iePPARγKO mice had higher plasma free FA levels and lower gastric inhibitory polypeptide levels than their wild-type (WT) littermates, and altered expression of incretin genes and lipid metabolism-associated genes in the intestinal epithelium. Gavage with the medium chain saturated FA-rich coconut oil did not result in differences between the two genotypes. Furthermore, the iePPARγKO mice did not exhibit defective lipid uptake and stomach emptying; however, their intestinal transit was more rapid than in WT mice. When fed a canola oil-rich diet for 4.5 months, iePPARγKO mice had higher body lean mass than the WT mice. We conclude that intestinal epithelium PPARγ is activated preferentially by long chain unsaturated FAs compared to medium chain saturated FAs. Furthermore, we hypothesize that the iePPARγKO phenotype originates from altered lipid metabolism and release in epithelial cells, as well as changes in intestinal motility.
Collapse
|
40
|
Abstract
In healthy individuals, metabolically quiescent T cells survey lymph nodes and peripheral tissues in search of cognate antigens. During infection, T cells that encounter cognate antigens are activated and - in a context-specific manner - proliferate and/or differentiate to become effector T cells. This process is accompanied by important changes in cellular metabolism (known as metabolic reprogramming). The magnitude and spectrum of metabolic reprogramming as it occurs in T cells in the context of acute infection ensure host survival. By contrast, altered T cell metabolism, and hence function, is also observed in various disease states, in which T cells actively contribute to pathology. In this Review, we introduce the idea that the spectrum of immune cell metabolic states can provide a basis for categorizing human diseases. Specifically, we first summarize the metabolic and interlinked signalling requirements of T cells responding to acute infection. We then discuss how metabolic reprogramming of T cells is linked to disease.
Collapse
|
41
|
Poursharifi P, Madiraju SRM, Prentki M. Monoacylglycerol signalling and ABHD6 in health and disease. Diabetes Obes Metab 2017; 19 Suppl 1:76-89. [PMID: 28880480 DOI: 10.1111/dom.13008] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 04/24/2017] [Accepted: 05/11/2017] [Indexed: 12/14/2022]
Abstract
Lipid metabolism dysregulation underlies chronic pathologies such as obesity, diabetes and cancer. Besides their role in structure and energy storage, lipids are also important signalling molecules regulating multiple biological functions. Thus, understanding the precise lipid metabolism enzymatic steps that are altered in some pathological conditions is helpful for designing better treatment strategies. Several monoacylglycerol (MAG) species are only recently being recognized as signalling lipid molecules in different tissues. Recent studies indicated the importance of the ubiquitously expressed serine hydrolase α/β-hydrolase domain 6 (ABHD6), which is a MAG hydrolase, in regulating signalling competent MAG in both central and peripheral tissues. The central and peripheral function of the endocannabinoid 2-arachidonoylglycerol, which is a 2-MAG, and its breakdown by both ABHD6 and classical MAG lipase has been well documented. ABHD6 and its substrate MAG appear to be involved in the regulation of various physiological and pathological processes including insulin secretion, adipose browning, food intake, neurotransmission, autoimmune disorders, neurological and metabolic diseases as well as cancer. Diverse cellular targets such as mammalian unc13-1 (Munc13-1), PPARs, GPR119 and CB1/2 receptors, for MAG-mediated signalling processes have been proposed in different cell types. The purpose of this review is to provide a comprehensive summary of the current state of knowledge regarding ABHD6/MAG signalling and its possible therapeutic implications.
Collapse
Affiliation(s)
- Pegah Poursharifi
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Sri Ramachandra Murthy Madiraju
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| | - Marc Prentki
- Departments of Nutrition, Biochemistry and Molecular Medicine, University of Montreal, and Montreal Diabetes Research Center, CRCHUM, Montreal, Canada
| |
Collapse
|
42
|
Brett E, Chung N, Leavitt WT, Momeni A, Longaker MT, Wan DC. A Review of Cell-Based Strategies for Soft Tissue Reconstruction. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:336-346. [PMID: 28372485 DOI: 10.1089/ten.teb.2016.0455] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Soft tissue reconstruction to restore volume to damaged or deficient tissue beneath the skin remains a challenging endeavor. Current techniques are centered around autologous fat transfer, or the use of synthetic substitutes, however, a great deal of scientific inquiry has been made into both the molecular mechanisms involved in, and limitations of, de novo adipogenesis, that is, the formation of new adipose tissue from precursor cells. To best comprehend these mechanisms, an understanding of defined markers for adipogenic differentiation, and knowledge of both commercially available and primary cell lines that enable in vitro and in vivo studies is necessary. We review the growth factors, proteins, cytokines, drugs, and molecular pathways that have shown promise in enhancing adipogenesis and vasculogenesis, in addition to the multitude of scaffolds that act as delivery vehicles to support these processes. While progress continues on these fronts, equally important is how researchers are optimizing clinically employed strategies such as autologous fat transfer through cell-based intervention, and the potential to augment this approach through isolation of preferentially adipogenic or angiogenic precursor subpopulations, which exists on the horizon. This review will highlight the novel molecular and synthetic modifications currently being studied for inducing adipose tissue regeneration on a cellular level, which will expand our arsenal of techniques for approaching soft tissue reconstruction.
Collapse
Affiliation(s)
- Elizabeth Brett
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Natalie Chung
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - William Tripp Leavitt
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Arash Momeni
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Michael T Longaker
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California.,2 Institute for Stem Cell Biology and Regenerative Medicine, Stanford University , Stanford, California
| | - Derrick C Wan
- 1 Hagey Laboratory for Pediatric Regenerative Medicine, Plastic and Reconstructive Surgery Division, Department of Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|
43
|
Commensal gut bacteria modulate phosphorylation-dependent PPARγ transcriptional activity in human intestinal epithelial cells. Sci Rep 2017; 7:43199. [PMID: 28266623 PMCID: PMC5339702 DOI: 10.1038/srep43199] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 01/20/2017] [Indexed: 02/08/2023] Open
Abstract
In healthy subjects, the intestinal microbiota interacts with the host’s epithelium, regulating gene expression to the benefit of both, host and microbiota. The underlying mechanisms remain poorly understood, however. Although many gut bacteria are not yet cultured, constantly growing culture collections have been established. We selected 57 representative commensal bacterial strains to study bacteria-host interactions, focusing on PPARγ, a key nuclear receptor in colonocytes linking metabolism and inflammation to the microbiota. Conditioned media (CM) were harvested from anaerobic cultures and assessed for their ability to modulate PPARγ using a reporter cell line. Activation of PPARγ transcriptional activity was linked to the presence of butyrate and propionate, two of the main metabolites of intestinal bacteria. Interestingly, some stimulatory CMs were devoid of these metabolites. A Prevotella and an Atopobium strain were chosen for further study, and shown to up-regulate two PPARγ-target genes, ANGPTL4 and ADRP. The molecular mechanisms of these activations involved the phosphorylation of PPARγ through ERK1/2. The responsible metabolites were shown to be heat sensitive but markedly diverged in size, emphasizing the diversity of bioactive compounds found in the intestine. Here we describe different mechanisms by which single intestinal bacteria can directly impact their host’s health through transcriptional regulation.
Collapse
|
44
|
Single-Nucleotide Polymorphism of PPARγ, a Protein at the Crossroads of Physiological and Pathological Processes. Int J Mol Sci 2017; 18:ijms18020361. [PMID: 28208577 PMCID: PMC5343896 DOI: 10.3390/ijms18020361] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/24/2017] [Accepted: 02/01/2017] [Indexed: 01/28/2023] Open
Abstract
Genome polymorphisms are responsible for phenotypic differences between humans and for individual susceptibility to genetic diseases and therapeutic responses. Non-synonymous single-nucleotide polymorphisms (nsSNPs) lead to protein variants with a change in the amino acid sequence that may affect the structure and/or function of the protein and may be utilized as efficient structural and functional markers of association to complex diseases. This study is focused on nsSNP variants of the ligand binding domain of PPARγ a nuclear receptor in the superfamily of ligand inducible transcription factors that play an important role in regulating lipid metabolism and in several processes ranging from cellular differentiation and development to carcinogenesis. Here we selected nine nsSNPs variants of the PPARγ ligand binding domain, V290M, R357A, R397C, F360L, P467L, Q286P, R288H, E324K, and E460K, expressed in cancer tissues and/or associated with partial lipodystrophy and insulin resistance. The effects of a single amino acid change on the thermodynamic stability of PPARγ, its spectral properties, and molecular dynamics have been investigated. The nsSNPs PPARγ variants show alteration of dynamics and tertiary contacts that impair the correct reciprocal positioning of helices 3 and 12, crucially important for PPARγ functioning.
Collapse
|
45
|
|
46
|
Zhang L, Xiao J, Xu J, Fu T, Cao Z, Zhu L, Chen HZ, Shen X, Jiang H, Zhang L. Crystal structure of FabZ-ACP complex reveals a dynamic seesaw-like catalytic mechanism of dehydratase in fatty acid biosynthesis. Cell Res 2016; 26:1330-1344. [PMID: 27874013 PMCID: PMC5143422 DOI: 10.1038/cr.2016.136] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 08/18/2016] [Accepted: 09/07/2016] [Indexed: 12/28/2022] Open
Abstract
Fatty acid biosynthesis (FAS) is a vital process in cells. Fatty acids are essential for cell assembly and cellular metabolism. Abnormal FAS directly correlates with cell growth delay and human diseases, such as metabolic syndromes and various cancers. The FAS system utilizes an acyl carrier protein (ACP) as a transporter to stabilize and shuttle the growing fatty acid chain throughout enzymatic modules for stepwise catalysis. Studying the interactions between enzymatic modules and ACP is, therefore, critical for understanding the biological function of the FAS system. However, the information remains unclear due to the high flexibility of ACP and its weak interaction with enzymatic modules. We present here a 2.55 Å crystal structure of type II FAS dehydratase FabZ in complex with holo-ACP, which exhibits a highly symmetrical FabZ hexamer-ACP3 stoichiometry with each ACP binding to a FabZ dimer subunit. Further structural analysis, together with biophysical and computational results, reveals a novel dynamic seesaw-like ACP binding and catalysis mechanism for the dehydratase module in the FAS system, which is regulated by a critical gatekeeper residue (Tyr100 in FabZ) that manipulates the movements of the β-sheet layer. These findings improve the general understanding of the dehydration process in the FAS system and will potentially facilitate drug and therapeutic design for diseases associated with abnormalities in FAS.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai, China
| | - Jianfeng Xiao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jianrong Xu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianran Fu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiwei Cao
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai, China
| | - Hong-Zhuan Chen
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai, China
| | - Xu Shen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hualiang Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liang Zhang
- Department of Pharmacology and Chemical Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai, China
| |
Collapse
|
47
|
Intestinal PPARγ signalling is required for sympathetic nervous system activation in response to caloric restriction. Sci Rep 2016; 6:36937. [PMID: 27853235 PMCID: PMC5113069 DOI: 10.1038/srep36937] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 10/05/2016] [Indexed: 02/08/2023] Open
Abstract
Nuclear receptor PPARγ has been proven to affect metabolism in multiple tissues, and has received considerable attention for its involvement in colon cancer and inflammatory disease. However, its role in intestinal metabolism has been largely ignored. To investigate this potential aspect of PPARγ function, we submitted intestinal epithelium-specific PPARγ knockout mice (iePPARγKO) to a two-week period of 25% caloric restriction (CR), following which iePPARγKO mice retained more fat than their wild type littermates. In attempting to explain this discrepancy, we analysed the liver, skeletal muscle, intestinal lipid trafficking, and the microbiome, none of which appeared to contribute to the adiposity phenotype. Interestingly, under conditions of CR, iePPARγKO mice failed to activate their sympathetic nervous system (SNS) and increase CR-specific locomotor activity. These KO mice also manifested a defective control of their body temperature, which was overly reduced. Furthermore, the white adipose tissue of iePPARγKO CR mice showed lower levels of both hormone-sensitive lipase, and its phosphorylated form. This would result from impaired SNS signalling and possibly cause reduced lipolysis. We conclude that intestinal epithelium PPARγ plays an essential role in increasing SNS activity under CR conditions, thereby contributing to energy mobilization during metabolically stressful episodes.
Collapse
|
48
|
Pereira VH, Marques F, Lages V, Pereira FG, Patchev A, Almeida OFX, Almeida-Palha J, Sousa N, Cerqueira JJ. Glucose intolerance after chronic stress is related with downregulated PPAR-γ in adipose tissue. Cardiovasc Diabetol 2016; 15:114. [PMID: 27538526 PMCID: PMC4990862 DOI: 10.1186/s12933-016-0433-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 08/06/2016] [Indexed: 01/09/2023] Open
Abstract
Background Chronic stress is associated with increased risk of glucose intolerance and cardiovascular diseases, albeit through undefined mechanisms. With the aim of gaining insights into the latter, this study examined the metabolic profile of young adult male rats that were exposed to chronic unpredictable stress. Methods Young adult male rats were submitted to 4 weeks of chronic unpredictable stress and allowed to recover for 5 weeks. An extensive analysis including of morphologic, biochemical and molecular parameters was carried out both after chronic unpredictable stress and after recovery from stress. Results After 28 days of chronic unpredictable stress (CUS) the animals submitted to this protocol displayed less weight gain than control animals. After 5 weeks of recovery the weight gain rebounded to similar values of controls. In addition, following CUS, fasting insulin levels were increased and were accompanied by signs of impaired glucose tolerance and elevated serum corticosteroid levels. This biochemical profile persisted into the post-stress recovery period, despite the restoration of baseline corticosteroid levels. The mRNA expression levels of peroxisome proliferator-activated receptor (PPAR)-γ and lipocalin-2 in white adipose tissue were, respectively, down- and up-regulated. Conclusions Reduction of PPAR-γ expression and generation of a pro-inflammatory environment by increased lipocalin-2 expression in white adipose tissue may contribute to stress-induced glucose intolerance.
Collapse
Affiliation(s)
- Vitor H Pereira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Vânia Lages
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Filipa G Pereira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | | | | | - Joana Almeida-Palha
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal. .,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| | - João J Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
49
|
Blanchard PG, Turcotte V, Côté M, Gélinas Y, Nilsson S, Olivecrona G, Deshaies Y, Festuccia WT. Peroxisome proliferator-activated receptor γ activation favours selective subcutaneous lipid deposition by coordinately regulating lipoprotein lipase modulators, fatty acid transporters and lipogenic enzymes. Acta Physiol (Oxf) 2016; 217:227-39. [PMID: 26918671 DOI: 10.1111/apha.12665] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/15/2015] [Accepted: 02/19/2016] [Indexed: 12/18/2022]
Abstract
AIM Peroxisome proliferator-activated receptor (PPAR) γ activation is associated with preferential lipoprotein lipase (LPL)-mediated fatty acid storage in peripheral subcutaneous fat depots. How PPARγ agonism acts upon the multi-level modulation of depot-specific lipid storage remains incompletely understood. METHODS We evaluated herein triglyceride-derived lipid incorporation into adipose tissue depots, LPL mass and activity, mRNA levels and content of proteins involved in the modulation of LPL activity and fatty acid transport, and the expression/activity of enzymes defining adipose tissue lipogenic potential in rats treated with the PPARγ ligand rosiglitazone (30 mg kg(-1) day(-1) , 23 days) after either a 10-h fasting period or a 17-h fast followed by 6 h of ad libitum refeeding. RESULTS Rosiglitazone stimulated lipid accretion in subcutaneous fat (SF) ~twofold and significantly reduced that of visceral fat (VF) to nearly half. PPARγ activation selectively increased LPL mass, activity and the expression of its chaperone LMF1 in SF. In VF, rosiglitazone had no effect on LPL activity and downregulated the mRNA levels of the transendothelial transporter GPIHBP1. Overexpression of lipid uptake and fatty acid transport proteins (FAT/CD36, FATP1 and FABP4) and stimulation of lipogenic enzyme activities (GPAT, AGPAT and DGAT) upon rosiglitazone treatment were of higher magnitude in SF. CONCLUSIONS Together these findings demonstrate that the depot-specific transcriptional control of LPL induced by PPARγ activation extends to its key interacting proteins and post-translational modulators to favour subcutaneous lipid storage.
Collapse
Affiliation(s)
- P. G. Blanchard
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - V. Turcotte
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - M. Côté
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - Y. Gélinas
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - S. Nilsson
- Department of Medical Biosciences/Physiological Chemistry; Umeå University; Umeå Sweden
| | - G. Olivecrona
- Department of Medical Biosciences/Physiological Chemistry; Umeå University; Umeå Sweden
| | - Y. Deshaies
- Department of Medicine; Faculty of Medicine; Quebec Heart and Lung Institute; Laval University; Quebec QC Canada
| | - W. T. Festuccia
- Department of Physiology and Biophysics; Institute of Biomedical Sciences; University of São Paulo; São Paulo Brazil
| |
Collapse
|
50
|
Jager J, Wang F, Fang B, Lim HW, Peed LC, Steger DJ, Won KJ, Kharitonenkov A, Adams AC, Lazar MA. The Nuclear Receptor Rev-erbα Regulates Adipose Tissue-specific FGF21 Signaling. J Biol Chem 2016; 291:10867-75. [PMID: 27002153 DOI: 10.1074/jbc.m116.719120] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Indexed: 01/14/2023] Open
Abstract
FGF21 is an atypical member of the FGF family that functions as a hormone to regulate carbohydrate and lipid metabolism. Here we demonstrate that the actions of FGF21 in mouse adipose tissue, but not in liver, are modulated by the nuclear receptor Rev-erbα, a potent transcriptional repressor. Interrogation of genes induced in the absence of Rev-erbα for Rev-erbα-binding sites identified βKlotho, an essential coreceptor for FGF21, as a direct target gene of Rev-erbα in white adipose tissue but not liver. Rev-erbα ablation led to the robust elevated expression of βKlotho. Consequently, the effects of FGF21 were markedly enhanced in the white adipose tissue of mice lacking Rev-erbα. A major Rev-erbα-controlled enhancer at the Klb locus was also bound by the adipocytic transcription factor peroxisome proliferator-activated receptor (PPAR) γ, which regulates its activity in the opposite direction. These findings establish Rev-erbα as a specific modulator of FGF21 signaling in adipose tissue.
Collapse
Affiliation(s)
- Jennifer Jager
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Fenfen Wang
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Bin Fang
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Hee-Woong Lim
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Lindsey C Peed
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David J Steger
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kyoung-Jae Won
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Alexei Kharitonenkov
- the Department of Chemistry, Indiana University Bloomington, Bloomington, Indiana 47405, and
| | - Andrew C Adams
- the Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana 46285
| | - Mitchell A Lazar
- From the Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Genetics, and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104,
| |
Collapse
|