1
|
Francis R, Kalyanaraman R, Boominathan V, Parthasarathy S, Chavaan A, Ansari IA, Ansari SA, Alkahtani HM, Chandran J, Tharumasivam SV. Piperine's potential in treating polycystic ovarian syndrome explored through in-silico docking. Sci Rep 2024; 14:21834. [PMID: 39294254 PMCID: PMC11411113 DOI: 10.1038/s41598-024-72800-6] [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: 05/03/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024] Open
Abstract
Polycystic Ovarian Syndrome (PCOS) is a multifaceted metabolic and hormonal condition that impacts women in their procreative ages, identified by ovarian dysfunction, hyperandrogenaemia overweight and insulin insensitivity. The piperine, an important alkaloid compound of black pepper has shown promise in modulating various physiological processes. In this work, employed computational docking studies to explore the potential of piperine as a treatment for PCOS. Utilizing computational methods, we analyzed the binding interactions between piperine and key molecular targets implicated in PCOS pathogenesis, including hyperandrogenism, and "oligomenorrhea. The network pharmacology analysis report found 988 PCOS-related genes, 108 hyperandrogenism-related genes, and 377 oligomenorrhea-related genes, and we finally shortlisted 5 common genes in PCOS, hyperandrogenism, and "oligomenorrhea": NR3C1, PPARG, FOS, CYP17A1, and H6PD. Our results reveal favorable binding affinities with PPARG (-8.34 Kcal/mol) and H6PD (-8.70 Kcal/mol) and interaction patterns, suggesting the potential of piperine to modulate these targets. Moreover, the reliability of the piperine-target interactions was revealed by molecular simulations studies. These findings support further experimental investigations to validate the therapeutic efficacy of piperine in PCOS management. The integration of computational approaches with experimental studies has the potential to lay the groundwork for the creation of new therapies specifically targeting PCOS and related endocrine disorders.
Collapse
Affiliation(s)
- Rahul Francis
- Department of Biotechnology, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Ramanathan Kalyanaraman
- Department of Biotechnology, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Vasuki Boominathan
- Department of Biotechnology, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | | | - Ashajyothi Chavaan
- Department of Studies in Biotechnology, Vijayanagar Sri Krishnadevarya University, Ballari, Karnataka, 583-105, India
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, 10125, Turin, Italy
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O Box 2457, 11451, Riyadh, Saudi Arabia.
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O Box 2457, 11451, Riyadh, Saudi Arabia
| | - Janani Chandran
- Department of Biotechnology, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Siva Vijayakumar Tharumasivam
- Department of Biotechnology, Srimad Andavan Arts and Science College (Autonomous), Affiliated to Bharathidasan University, Tiruchirappalli, Tamil Nadu, India.
- Department of Biotechnology Engineering, School of Engineering and Technology, Dhanalakshmi Srinivasan University, Samayapuram, Trichy, Tamil Nadu, India.
| |
Collapse
|
2
|
Xie Z, Huang M, Xu W, Liu F, Huang D. USP18 Curbs the Progression of Metabolic Hypertension by Suppressing JAK/STAT Pathway. Cardiovasc Toxicol 2024; 24:576-586. [PMID: 38691302 DOI: 10.1007/s12012-024-09860-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 04/13/2024] [Indexed: 05/03/2024]
Abstract
Hypertension is a pathological state of the metabolic syndrome that increases the risk of cardiovascular disease. Managing hypertension is challenging, and we aimed to identify the pathogenic factors and discern therapeutic targets for metabolic hypertension (MHR). An MHR rat model was established with the combined treatment of a high-sugar, high-fat diet and ethanol. Histopathological observations were performed using hematoxylin-eosin and Sirius Red staining. Transcriptome sequencing was performed to screen differentially expressed genes. The role of ubiquitin-specific protease 18 (USP18) in the proliferation, apoptosis, and oxidative stress of HUVECs was explored using Cell Counting Kit-8, flow cytometry, and enzyme-linked immunosorbent assays. Moreover, USP18 downstream signaling pathways in MHR were screened, and the effects of USP18 on these signaling pathways were investigated by western blotting. In the MHR model, total cholesterol and low-density lipoprotein levels increased, while high-density lipoprotein levels decreased. Moreover, high vessel thickness and percentage of collagen were noted along with increased malondialdehyde, decreased superoxide dismutase and catalase levels. The staining results showed that the MHR model exhibited an irregular aortic intima and disordered smooth muscle cells. There were 78 differentially expressed genes in the MHR model, and seven hub genes, including USP18, were identified. USP18 overexpression facilitated proliferation and reduced apoptosis and oxidative stress in HUVECs treated with Ang in vitro. In addition, the JAK/STAT pathway was identified as a USP18 downstream signaling pathway, and USP18 overexpression inhibited the expression of JAK/STAT pathway-related proteins. Conclusively, USP18 restrained MHR progression by promoting cell proliferation, reversing apoptosis and oxidative stress, and suppressing the JAK/STAT pathway.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Rats
- Apoptosis/drug effects
- Blood Pressure/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Disease Progression
- Gene Expression Regulation
- Human Umbilical Vein Endothelial Cells/metabolism
- Human Umbilical Vein Endothelial Cells/pathology
- Human Umbilical Vein Endothelial Cells/drug effects
- Human Umbilical Vein Endothelial Cells/enzymology
- Hypertension/metabolism
- Hypertension/physiopathology
- Hypertension/pathology
- Hypertension/enzymology
- Janus Kinases/metabolism
- Metabolic Syndrome/metabolism
- Metabolic Syndrome/pathology
- Metabolic Syndrome/enzymology
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/enzymology
- Oxidative Stress/drug effects
- Rats, Sprague-Dawley
- Signal Transduction
- STAT Transcription Factors/metabolism
- Ubiquitin Thiolesterase/metabolism
- Ubiquitin Thiolesterase/genetics
- Vascular Remodeling/drug effects
Collapse
Affiliation(s)
- Zhihong Xie
- Department of Cardiology, Ganzhou People's Hospital, 16 Meiguan Dadao, Zhanggong District, Ganzhou, 341000, Jiangxi, China.
| | - Mingshan Huang
- Department of Cardiology, Ganzhou People's Hospital, 16 Meiguan Dadao, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Wang Xu
- Department of Cardiology, Ganzhou People's Hospital, 16 Meiguan Dadao, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Fuwei Liu
- Department of Cardiology, Ganzhou People's Hospital, 16 Meiguan Dadao, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| | - Donghua Huang
- Department of Cardiology, Ganzhou People's Hospital, 16 Meiguan Dadao, Zhanggong District, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
3
|
Xie L, Huang B, Zhao X, Zhu N. Exploring the mechanisms underlying effects of bisphenol a on cardiovascular disease by network toxicology and molecular docking. Heliyon 2024; 10:e31473. [PMID: 38813174 PMCID: PMC11133888 DOI: 10.1016/j.heliyon.2024.e31473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/15/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024] Open
Abstract
Background Globally, cardiovascular disease (CVD) has emerged as a leading cause of mortality. Bisphenol A (BPA), recognized as one of the most prevalent and widely distributed endocrine-disrupting chemicals (EDCs), has been consistently linked to the progression of CVD. This research centers on unraveling the molecular mechanisms responsible for the toxic effects of BPA exposure on CVD. Key targets and pathways involved in action of BPA on CVD were investigated by network toxicology. Binding abilities of BPA to core targets were evaluated by molecular docking. Methods and results Based on information retrieved from ChEMBL, DrugBank, and OMIM databases, a total of 27 potential targets were found to be associated with the influence of BPA on CVD. Furthermore, the STRING and Cytoscape software were employed to identify three central genes-ESR1, PPARG, and PTGS2-and to construct both the protein-protein interaction network and an interaction diagram of potential targets. Gene ontology (GO) and KEGG (Kyoto Encyclopedia of Genes and Genomes, KEGG) pathway enrichment analyses via WebGestalt revealed key biological processes (BP), cellular components (CC), molecular functions (MF), and pathways, such as the calcium signaling pathway, inflammatory mediator regulation of TRP channels, gap junction, adrenergic signaling in cardiomyocytes, cGMP-PKG signaling pathway, and cAMP signaling pathway, predominantly involved in BPA-induced CVD toxicity. By using molecular docking investigations, it proved that BPA binds to ESR1, PPARG, and PTGS2 steadily and strongly. Conclusion This study not only establishes a theoretical framework for understanding the molecular toxicity mechanism of BPA in cardiovascular disease (CVD) but also introduces an innovative network toxicology approach to methodically investigate the influence of environmental contaminants on CVD. This methodology sets the stage for drug discovery efforts targeting CVD linked to exposure to endocrine-disrupting chemicals (EDCs).
Collapse
Affiliation(s)
- Lina Xie
- Department of Neurosurgery, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, China
| | - Xuyong Zhao
- Department of Cardiology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| | - Ning Zhu
- Department of Cardiology, The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, China
| |
Collapse
|
4
|
Yu P, Wang W, Guo W, Cheng L, Wan Z, Cheng Y, Shen Y, Xu F. Pioglitazone-Enhanced Brown Fat Whitening Contributes to Weight Gain in Diet-Induced Obese Mice. Exp Clin Endocrinol Diabetes 2023; 131:595-604. [PMID: 37729949 DOI: 10.1055/a-2178-9113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
INTRODUCTION Pioglitazone is an insulin sensitizer used for the treatment of type 2 diabetes mellitus (T2DM) by activating peroxisome proliferator-activated receptor gamma. This study aimed to investigate the effects of pioglitazone on white adipose tissue (WAT) and brown adipose tissue (BAT) in diet-induced obese (DIO) mice. METHODS C57BL/6 mice were treated with pioglitazone (30 mg/kg/day) for 4 weeks after a 16-week high-fat diet (HFD) challenge. Body weight gain, body fat mass, energy intake, and glucose homeostasis were measured during or after the treatment. Histopathology was observed by hematoxylin and eosin, oil red O, immunohistochemistry, and immunofluorescence staining. Expression of thermogenic and mitochondrial biogenesis-related genes was detected by quantitative real-time PCR and western blotting. RESULTS After 4-week pioglitazone treatment, the fasting blood glucose levels, glucose tolerance, and insulin sensitivity were significantly improved, but the body weight gain and fat mass were increased in DIO mice. Compared with the HFD group, pioglitazone did not significantly affect the weights of liver and WAT in both subcutaneous and epididymal regions. Unexpectedly, the weight of BAT was increased after pioglitazone treatment. Histological staining revealed that pioglitazone ameliorated hepatic steatosis, reduced the adipocyte size in WAT, but increased the adipocyte size in BAT. CONCLUSION Though pioglitazone can promote lipolysis, thermogenesis, and mitochondrial function in WAT, it leads to impaired thermogenesis, and mitochondrial dysfunction in BAT. In conclusion, pioglitazone could promote the browning of WAT but led to the whitening of BAT; the latter might be a new potential mechanism of pioglitazone-induced weight gain during T2DM treatment.
Collapse
Affiliation(s)
- Piaojian Yu
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong Province, China
- Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University
| | - Wei Wang
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong Province, China
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Wanrong Guo
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong Province, China
| | - Lidan Cheng
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong Province, China
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhiping Wan
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong Province, China
- Department of Infectious Diseases, the Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yanglei Cheng
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yunfeng Shen
- Department of Endocrinology and Metabolism, the Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang, Jiangxi Province, China
| | - Fen Xu
- Department of Endocrinology and Metabolism, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, Guangdong Province, China
- Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity Research, The Third Affiliated Hospital of Sun Yat-sen University
| |
Collapse
|
5
|
Li Y, Long J, Li L, Yu Z, Liang Y, Hou B, Xiang L, Niu X. Pioglitazone protects PC12 cells against oxidative stress injury: An in vitro study of its antiapoptotic effects via the PPARγ pathway. Exp Ther Med 2023; 26:522. [PMID: 37854503 PMCID: PMC10580242 DOI: 10.3892/etm.2023.12221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 01/20/2023] [Indexed: 10/20/2023] Open
Abstract
To the best of our knowledge, the role of peroxisome proliferator-activated receptor γ (PPARγ) in oxidative stress-induced PC12 cell damage is unknown. Using a PC12 cell model with H2O2 treatment, the present study investigated the expression levels of apoptosis-related genes and neuronal apoptosis after oxidative stress injury. The present study further investigated the protective effect and mechanism of pioglitazone, a PPARγ agonist. PC12 cells treated with H2O2 were used as a model of oxidative stress injury. An MTT assay and flow cytometry were used to detect the effect of H2O2 on PC12 cell viability and the protective effect of pioglitazone. A TUNEL assay was used to detect neuronal apoptosis. The expression levels of PPARγ, Bax, Bcl-2 and caspase-3 were examined by reverse transcription-quantitative PCR and western blotting. H2O2 reduced PC12 cell viability in a dose- and time-dependent manner. H2O2 significantly upregulated the protein expression levels of Bax and the cleaved caspase-3/caspase-3 ratio (P<0.01), decreased the protein expression levels of Bcl-2 (P<0.01), and increased the apoptosis rate of PC12 cells. Pioglitazone significantly reduced the protein expression levels of Bax and the cleaved caspase-3/caspase-3 ratio (P<0.01), increased the expression levels of Bcl-2 (P<0.01), decreased the Bax/Bcl-2 expression ratio (P<0.01) and increased the viability of H2O2-damaged PC12 cells in a dose-dependent manner. Treatment with the PPARγ antagonist GW9662 or PPARγ small interfering RNA counteracted the protective effect of pioglitazone on PC12 cells to different extents (P<0.01). Therefore, the present study reported the role of PPARγ in protecting PC12 cells against oxidative stress injury, which may lead to novel therapeutic approaches for neurodegenerative diseases.
Collapse
Affiliation(s)
- Yali Li
- Department of Rehabilitation, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong 264200, P.R. China
- Department of Rehabilitation Medicine, Affiliated Hospital of Weifang Medical University, Weifang, Shandong 261000, P.R. China
| | - Jun Long
- Department of Rehabilitation, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Libo Li
- Department of Rehabilitation, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Ziyao Yu
- College of Sports and Health, Shandong Sport University, Jinan, Shandong 250000, P.R. China
| | - Yanjing Liang
- Department of Rehabilitation, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Bin Hou
- Department of Rehabilitation, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong 264200, P.R. China
| | - Li Xiang
- Department of Neurology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| | - Xiaolin Niu
- Department of Cardiology, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi 710004, P.R. China
| |
Collapse
|
6
|
Said MA, Nafeh NY, Abdallah HA. Spexin alleviates hypertension, hyperuricaemia, dyslipidemia and insulin resistance in high fructose diet induced metabolic syndrome in rats via enhancing PPAR-ɣ and AMPK and inhibiting IL-6 and TNF-α. Arch Physiol Biochem 2023; 129:1111-1116. [PMID: 33721543 DOI: 10.1080/13813455.2021.1899242] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/18/2022]
Abstract
Spexin is a novel peptide implicated in obesity and energy homeostasis. The objective of the current study was to evaluate the effect of spexin on blood pressure, insulin resistance, and dyslipidemia in rats with metabolic syndrome (MS) induced by high-fructose diet (HFD) and the possible underlying mechanism. Forty adult male rats were randomly assigned into four equal groups; Control, Spexin, HFD and HFD + spexin. Induction of the MS with HFD was associated with increased body mass index, elevated blood pressure, blood glucose, insulin, uric acid, advanced glycation end products and insulin resistance, interlekin-6, tumour necrosis factor-alpha together with dyslipidemia, low-serum spexin, peroxisome proliferator-activated receptors-gamma (PPAR-ɣ) and adenosine monophosphate-activated protein kinase (AMPK). Spexin attenuated MS-induced deleterious effects which can be attributed to activation of PPAR-ɣ and AMPK as well as inhibiting inflammation. These findings indicate that spexin could be a beneficial complementary agent for metabolic syndrome treatment.
Collapse
Affiliation(s)
- Mona A Said
- Physiology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Naglaa Y Nafeh
- Physiology Department, Faculty of Medicine, Benha University, Benha, Egypt
| | - Hend A Abdallah
- Physiology Department, Faculty of Medicine, Benha University, Benha, Egypt
| |
Collapse
|
7
|
Elkhattabi L, Zouhdi S, Moussetad F, Kettani A, Barakat A, Saile R. Molecular docking analysis of PPARγ with phytochemicals from Moroccan medicinal plants. Bioinformation 2023; 19:795-806. [PMID: 37901293 PMCID: PMC10605085 DOI: 10.6026/97320630019795] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 10/31/2023] Open
Abstract
PPARγ agonists play a crucial role in regulating metabolic homeostasis for treating type-2 diabetes (T2D). Due to the adverse side effects associated with thiazolidinediones, a class of PPARγ agonists, there is a growing interest in identifying natural compounds from medicinal plants that have the potential to bind PPARγ. In this study, we extensively investigated Moroccan phytochemicals using computational structure-based screening with the crystal structure of the PPARγ ligand-binding domain (PDB ID: 7awc) to discover novel phytochemicals targeting PPARγ. The docking results of 540 Moroccan phytochemicals were integrated into online databases for further exploitation through in-depth studies. Drug-likeness analysis was performed to assess the phytochemicals drug-like properties. Two promising phytochemicals, 3,4-dicaffeoylquinic acid and Chlorogenic acid, were identified, both exhibiting high docking affinity and unique binding site interactions compared to the established PPARγ full agonist, rosiglitazone. Molecular dynamics simulations of 100 ns were conducted to examine the stability of the complexes formed by both compounds within the PPARγ active site, and their dynamic behavior was compared to the reference structure of PPARγ alone and with rosiglitazone. Binding free energy calculations demonstrated that 3,4-dicaffeoylquinic acid and Chlorogenic acid exhibited higher binding free energy than the reference agonist, suggesting their potential as candidates for experimental validation in future drug discovery efforts targeting PPARγ for the treatment of T2D and metabolic syndrome.
Collapse
Affiliation(s)
- Lamiae Elkhattabi
- />Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Morocco
- />Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca
| | - Salwa Zouhdi
- />Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Morocco
| | - Fairouz Moussetad
- />Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Morocco
| | - Anass Kettani
- />Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Morocco
| | - Abdelhamid Barakat
- />Laboratory of Genomics and Human Genetics, Institut Pasteur du Maroc, Casablanca
| | - Rachid Saile
- />Laboratory of Biology and Health, Faculty of Sciences Ben M'Sik, Hassan II University of Casablanca, Morocco
| |
Collapse
|
8
|
Lan Z, Zhang K, He J, Kang Q, Meng W, Wang S. Pectolinarigenin shows lipid-lowering effects by inhibiting fatty acid biosynthesis in vitro and in vivo. Phytother Res 2023; 37:913-925. [PMID: 36415143 DOI: 10.1002/ptr.7679] [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: 08/05/2022] [Revised: 10/21/2022] [Accepted: 11/06/2022] [Indexed: 11/24/2022]
Abstract
Pectolinarigenin is the main flavonoid compound and presents in Linaria vulgaris and Cirsium chanroenicum. In this study, RNA sequencing (RNA-seq) was applied to dissect the effect of pectolinarigenin on the transcriptome changes in the high lipid Huh-7 cells induced by oleic acid. RNA-seq results revealed that 15 pathways enriched by downregulated genes are associated with cell metabolism including cholesterol metabolism, glycerophospholipid metabolism, steroid biosynthesis, steroid hormone biosynthesis, fatty acid biosynthesis, etc. Moreover, 13 key genes related to lipid metabolism were selected. Among them, PPARG coactivator 1 beta (PPARGC1B) and carnitine palmitoyltransferase 1A (CPT1A) were found to be upregulated, solute carrier family 27 member 1(SLC27A1), acetyl-CoA carboxylase alpha (ACACA), fatty-acid synthase (FASN), 3-Hydroxy-3-Methylglutaryl-CoA Reductase (HMGCR), etc. were found to be downregulated. Glycolysis/gluconeogenesis, steroid hormone biosynthesis, and fatty acid biosynthesis were all significantly downregulated, according to gene set variation analysis and gene set enrichment analysis. Besides, protein levels of FASN, ACACA, and SLC27A1 were all decreased, whereas PPARγ and CPT1A were increased. Docking models showed that PPARγ may be a target for pectolinarigenin. Furthermore, pectolinarigenin reduced serum TG and hepatic TG, and improved insulin sensitivity in vivo. Our findings suggest that pectolinarigenin may target PPARγ and prevent fatty acid biosynthesis.
Collapse
Affiliation(s)
- Zhou Lan
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Kun Zhang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Jianhui He
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Qiong Kang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Wei Meng
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| | - Songhua Wang
- Jiangxi Provincial Key Laboratory of Drug Design and Evaluation, Jiangxi Key Laboratory of Organic Chemistry, Jiangxi Science and Technology Normal University, Nanchang, China
| |
Collapse
|
9
|
Fan Y, Xu F, Wang R, He J. Lysine 222 in PPAR γ1 functions as the key site of MuRF2-mediated ubiquitination modification. Sci Rep 2023; 13:1999. [PMID: 36737649 PMCID: PMC9898238 DOI: 10.1038/s41598-023-28905-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPAR γ) plays key roles in the development, physiology, reproduction, and homeostasis of organisms. Its expression and activity are regulated by various posttranslational modifications. We previously reported that E3 ubiquitin ligase muscle ring finger protein 2 (MuRF2) inhibits cardiac PPAR γ1 protein level and activity, eventually protects heart from diabetic cardiomyopathy; furthermore, by GST-pulldown assay, we found that MuRF2 modifies PPAR γ1 via poly-ubiquitination and accelerates PPAR γ1 proteasomal degradation. However, the key ubiquitination site on PPAR γ that MuRF2 targets for remains unclear. In the present study, we demonstrate that lysine site 222 is the receptor of MuRF2-mediated PPAR γ1 ubiquitination modification, using prediction of computational models, immunoprecipitation, ubiquitination assays, cycloheximide chasing assay and RT-qPCR. Our findings elucidated the underlying details of MuRF2 prevents heart from diabetic cardiomyopathy through the PPAR γ1 regulatory pathway.
Collapse
Affiliation(s)
- Yucheng Fan
- Department of Pathology, The First People's Hospital of Shizuishan, Affiliated to Ningxia Medical University, Shizuishan, China
| | - Fangjing Xu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Rui Wang
- School of Basic Medical Sciences , Ningxia Medical University, Yinchuan, China
| | - Jun He
- Department of Cardiovascular Internal Medicine, General Hospital of Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
10
|
Zahr T, Liu L, Chan M, Zhou Q, Cai B, He Y, Aaron N, Accili D, Sun L, Qiang L. PPARγ (Peroxisome Proliferator-Activated Receptor γ) Deacetylation Suppresses Aging-Associated Atherosclerosis and Hypercholesterolemia. Arterioscler Thromb Vasc Biol 2023; 43:30-44. [PMID: 36453279 PMCID: PMC9917767 DOI: 10.1161/atvbaha.122.318061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Atherosclerosis is a medical urgency manifesting at the onset of hypercholesterolemia and is associated with aging. Activation of PPARγ (peroxisome proliferator-activated receptor γ) counteracts metabolic dysfunction influenced by aging, and its deacetylation displays an atheroprotective property. Despite the marked increase of PPARγ acetylation during aging, it is unknown whether PPARγ acetylation is a pathogenic contributor to aging-associated atherosclerosis. METHODS Mice with constitutive deacetylation-mimetic PPARγ mutations on lysine residues K268 and K293 (2KR) in an LDL (low-density lipoprotein)-receptor knockout (Ldlr-/-) background (2KR:Ldlr-/-) were aged for 18 months on a standard laboratory diet to examine the cardiometabolic phenotype, which was confirmed in Western-type diet-fed 2KR:Ldlr+/- mice. Whole-liver RNA-sequencing and in vitro studies in bone marrow-derived macrophages were conducted to decipher the mechanism. RESULTS In contrast to severe atherosclerosis in WT:Ldlr-/- mice, aged 2KR:Ldlr-/- mice developed little to no plaque, which was underlain by a significantly improved plasma lipid profile, with particular reductions in circulating LDL. The protection from hypercholesterolemia was recapitulated in Western-type diet-fed 2KR:Ldlr+/- mice. Liver RNA-sequencing analysis revealed suppression of liver inflammation rather than changes in cholesterol metabolism. This anti-inflammatory effect of 2KR was attributed to polarized M2 activation of macrophages. Additionally, the upregulation of core circadian component Bmal1 (brain and muscle ARNT-like 1), perceived to be involved in anti-inflammatory immunity, was observed in the liver and bone marrow-derived macrophages. CONCLUSIONS PPARγ deacetylation in mice prevents the development of aging-associated atherosclerosis and hypercholesterolemia, in association with the anti-inflammatory phenotype of 2KR macrophages.
Collapse
Affiliation(s)
- Tarik Zahr
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, New York, USA
| | - Longhua Liu
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Michelle Chan
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
| | - Qiuzhong Zhou
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ying He
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Nicole Aaron
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Molecular Pharmacology and Therapeutics, Columbia University, New York, New York, USA
| | - Domenico Accili
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Medicine, Columbia University, New York, New York, USA
| | - Lei Sun
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Medical School, Singapore
| | - Li Qiang
- Department of Naomi Berrie Diabetes Center, Columbia University, New York, New York, USA
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| |
Collapse
|
11
|
Okagu IU, Ezeorba TPC, Aham EC, Aguchem RN, Nechi RN. Recent findings on the cellular and molecular mechanisms of action of novel food-derived antihypertensive peptides. FOOD CHEMISTRY. MOLECULAR SCIENCES 2022; 4:100078. [PMID: 35415696 PMCID: PMC8991738 DOI: 10.1016/j.fochms.2022.100078] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 01/07/2022] [Accepted: 01/21/2022] [Indexed: 12/14/2022]
Abstract
Hypertension has remained a silent-killer. Novel peptides recently isolated from food proteins. Molecular mechanism of blood pressure-lowering: renin and ACE-inhibition, and beyond. Proposed molecular mechanisms for future research. Novel peptides are excellent candidates for nutraceutical development.
Hypertension impacts negatively on the quality of life of sufferers, and complications associated with uncontrolled hypertension are life-threatening. Hence, many research efforts are exploring the antihypertensive properties of bioactive peptides derived from food proteins using in vitro ACE-inhibitory assay, experimentally-induced and spontaneous hypertensive rats, normotensive and hypertensive human models. In this study, the cellular and molecular mechanisms of blood pressure-lowering properties of novel peptides reported in recent studies (2015-July 30, 2021) were discussed. In addition to common mechanisms such as the inhibition of angiotensin I-converting enzyme (ACE) and renin activities, recently recognized mechanisms through which bioactive peptides exert their antihypertensive properties including the induction of vasodilation via upregulation of cyclo-oxygenase (COX) and prostaglandin receptor and endothelial nitric oxide synthase expression and L-type Ca2+ channel blockade were presented. Similarly, emerging mechanisms of blood pressure-lowering by bioactive peptides such as modulation of inflammation (TNF-α, and other cytokines signaling), oxidative stress (Keap-1/Nrf2/ARE/HO-1 and related signaling pathways), PPAR-γ/caspase3/MAPK signaling pathways and inhibition of lipid accumulation were discussed. The review also highlighted factors that influence the antihypertensive properties of peptides such as method of hydrolysis (type and number of enzymes, and chemical used for hydrolysis, and microbial fermentation), and amino acid sequence and chain length of peptides.
Collapse
Affiliation(s)
- Innocent U Okagu
- Department of Biochemistry, University of Nigeria, Nsukka 410001, Nigeria
| | | | - Emmanuel C Aham
- Department of Biochemistry, University of Nigeria, Nsukka 410001, Nigeria
| | - Rita N Aguchem
- Department of Biochemistry, University of Nigeria, Nsukka 410001, Nigeria
| | - Regina N Nechi
- Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka 410001, Nigeria
| |
Collapse
|
12
|
Wei M, Guo M, Meng X, Li L, Wang H, Zhang M, Bei Y. PPARγ Mediates the Cardioprotective Roles of Danlou Tablet After Acute Myocardial Ischemia-Reperfusion Injury. Front Cardiovasc Med 2022; 9:858909. [PMID: 35402529 PMCID: PMC8990898 DOI: 10.3389/fcvm.2022.858909] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 02/28/2022] [Indexed: 02/02/2023] Open
Abstract
Ischemic heart disease is one of the biggest threats to human life in the world. Reperfusion therapy is an effective strategy to reduce infarct size and ischemic injury. However, reperfusion process may cause secondary myocardial injury which is defined as ischemia-reperfusion injury (IRI). Exploring potential therapeutic strategy to attenuate IRI is extremely important. Danlou tablet (Dan), a Chinese herbal compound consisting of ten herbs, has been identified to be protective for the heart. However, the mechanism of Dan-induced cardioprotection after acute reperfusion was unelucidated. In this study, to investigate the role and mechanism of Dan in myocardial IRI, we performed acute IRI modeling in mice and oxygen-glucose deprivation–reperfusion (OGD/R)-induced apoptosis in primary neonatal rat cardiomyocytes (NRCMs). We found that Dan had protective effect against acute IRI in mice, as evidenced by reduced infarct size, TUNEL-positive cardiomyocytes (CMs), and Bax/Bcl2 ratio and cleaved-caspase 3/caspase 3 ratio in vivo. Meanwhile, Dan inhibited OGD/R-induced apoptosis of NRCMs in vitro. Mechanistically, Dan could activate proliferator-activated receptor gamma (PPARγ) in both IRI hearts and OGD/R-stressed NRCMs, while inhibition of PPARγ attenuated the protective effect of Dan against IRI in vivo and OGD/R-induced CM apoptosis in vitro. These data reveal that Dan attenuates acute myocardial IRI and CM apoptosis through activating PPARγ. Our findings may extend the knowledge of Chinese medicine and provide potential strategy for the precise treatment of ischemic heart diseases.
Collapse
Affiliation(s)
- Meng Wei
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Mengying Guo
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Xinxiu Meng
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Lin Li
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
| | - Hongyun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
- *Correspondence: Hongyun Wang
| | - Mingxue Zhang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
- Mingxue Zhang
| | - Yihua Bei
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong, China
- Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai, China
- Yihua Bei
| |
Collapse
|
13
|
Abstract
This review highlights molecular mechanisms of anti-inflammatory and protective
effects of the nuclear transcription factor, peroxisome proliferator-activated
receptor γ (PPARγ) in vascular tissue. PPARγ is an ubiquitously expressed
nuclear factor, and well-studied in adipose tissue and inflammatory cells.
Additionally, beneficial effects of vascular PPARγ’s on atherosclerosis and
vascular remodeling/dysfunction have been reported although the detailed
mechanism remains to be completely elucidated. Clinical and basic studies have
shown that the synthetic PPARγ ligands, thiazolidinediones (TZDs), have
protective effects against cardiovascular diseases such as atherosclerosis.
Recent studies utilizing genetic tools suggested that those protective effects
of TZDs on cardiovascular diseases are not due to a consequence of improvement
of insulin resistance, but may be due to a direct effect on PPARγ’s in vascular
endothelial and smooth muscle cells. In this review, we discuss proposed
mechanisms by which the vascular PPARγ regulates vascular inflammation and
remodeling/dysfunction especially in smooth muscle cells.
Collapse
Affiliation(s)
- Masashi Mukohda
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| | - Hiroshi Ozaki
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Ehime 794-8555, Japan
| |
Collapse
|
14
|
Li J, Yang T, Sha Z, Tang H, Hua X, Wang L, Wang Z, Gao Z, Sluijter JPG, Rowe GC, Das S, Yang L, Xiao J. Angiotensin II-induced muscle atrophy via PPARγ suppression is mediated by miR-29b. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 23:743-756. [PMID: 33614226 PMCID: PMC7868689 DOI: 10.1016/j.omtn.2020.12.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 12/19/2020] [Indexed: 12/20/2022]
Abstract
The activation of the renin-angiotensin system (RAS) induced by increased angiotensin II (AngII) levels has been implicated in muscle atrophy, which is involved in the pathogenesis of congestive heart failure. Although peroxisome proliferator-activated receptor gamma (PPARγ) activation can suppress RAS, the exact role of PPARγ in AngII-induced muscle atrophy is unclear. Here we identified PPARγ as a negative regulator of miR-29b, a microRNA that is able to promote multiple types of muscle atrophy. Suppression of miR-29b could prevent AngII-induced muscle atrophy both in vitro and in vivo. IGF1, PI3K(p85α), and Yin Yang 1 (YY1) were identified as target genes of miR-29b, and overexpression of these targets could rescue AngII-induced muscle atrophy. Importantly, inhibition of PPARγ was sufficient to induce muscle atrophy, while PPARγ overexpression could attenuate that. These data indicate that the PPARγ/miR-29b axis mediates AngII-induced muscle atrophy, and increasing PPARγ or inhibiting miR-29b represents a promising approach to counteract AngII-induced muscle atrophy.
Collapse
Affiliation(s)
- Jin Li
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Tingting Yang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Zhao Sha
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Haifei Tang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xuejiao Hua
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Lijun Wang
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Zitong Wang
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Ziyu Gao
- Department of Pathophysiology, Basic Medical Science, Harbin Medical University, Harbin 150081, China
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, Utrecht 3508GA, the Netherlands.,UMC Utrecht Regenerative Medicine Center, University Medical Center, Utrecht University, Utrecht 3508GA, the Netherlands
| | - Glenn C Rowe
- Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Saumya Das
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02215, USA
| | - Liming Yang
- Department of Pathophysiology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Junjie Xiao
- Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
15
|
Yao Y, Xu F, Ju X, Li Z, Wang L. Lipid-Lowering Effects and Intestinal Transport of Polyphenol Extract from Digested Buckwheat in Caco-2/HepG2 Coculture Models. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:4205-4214. [PMID: 32141744 DOI: 10.1021/acs.jafc.0c00321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Polyphenol extracts derived from gastrointestinal digestates of buckwheat (Fagopyrum Mill) were studied for their intestinal transport and lipid-lowering effects in Caco-2/HepG2 coculture models. The relative amounts of all phenolic compounds throughout the digestion and intestinal absorption process were determined by UHPLC-Q-Orbitrap mass spectrometry. The digestible and easily transported phenolic compounds in buckwheat extract were identified. Herein, four main phenolic compounds and their metabolites were found on both the apical and basolateral sides of the Caco-2 cell transwell model. The transepithelial transport rates in the Caco-2 cell monolayer were scoparone (0.97) > hydroxycinnamic acid (0.40) > rutin (0.23) > quercetin (0.20). The main metabolism of hydroxycinnamic acid, quercetin, and scoparone in transepithelial transport was found to be methylation. Furthermore, results indicated that triglyceride, low-density lipoprotein cholesterol, total cholesterol, aspartate aminotransferase, and alanine aminotransferase levels in HepG2 cells on the basolateral side of coculture models can be suppressed by 53.64, 23.44, 36.49, 27.98, and 77.42% compared to the oleic acid-induced group (p < 0.05). In addition, the mRNA expression of Fabp4 relative to the control was found to be significantly upregulated (85.82 ± 10.64 to 355.18 ± 65.83%) by the easily transported buckwheat polyphenol components in HepG2 cells (p < 0.01).
Collapse
Affiliation(s)
- Yijun Yao
- National Engineering Laboratory for Cereal Fermentation Technology, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, People's Republic of China
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, No. 3 Wenyuan Road, Nanjing, Jiangsu 210023, People's Republic of China
| | - Feiran Xu
- National Engineering Laboratory for Cereal Fermentation Technology, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, People's Republic of China
| | - Xingrong Ju
- National Engineering Laboratory for Cereal Fermentation Technology, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, 1800 Lihu Road, Wuxi 214122, Jiangsu, People's Republic of China
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, No. 3 Wenyuan Road, Nanjing, Jiangsu 210023, People's Republic of China
| | - Zhifang Li
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, No. 3 Wenyuan Road, Nanjing, Jiangsu 210023, People's Republic of China
| | - Lifeng Wang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, No. 3 Wenyuan Road, Nanjing, Jiangsu 210023, People's Republic of China
| |
Collapse
|
16
|
Algandaby MM. Crocin prevents metabolic syndrome in rats via enhancing PPAR-gamma and AMPK. Saudi J Biol Sci 2020; 27:1310-1316. [PMID: 32346340 PMCID: PMC7182989 DOI: 10.1016/j.sjbs.2020.01.004] [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] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/19/2019] [Accepted: 01/06/2020] [Indexed: 12/29/2022] Open
Abstract
Metabolic syndrome (Mets) is a major health hazard. The syndrome is strongly linked to cardiovascular disease. The objective of the current study was to address whether or not crocin could protect against experimentally-induced MetS in rats as well as the possible underlying mechanisms. Animals were allocated into 4 groups. The first one served as control and was kept on regular food pellets and drinking water. The other three groups were subjected to experimental MetS. Induction of MetS was achieved by keeping rats on food pellets containing 3% NaCl; and drinking water enriched with 10% fructose. The first and second groups were daily gavaged with saline. The third and fourth groups were daily administered crocin 5 and 10 mg/kg, respectively. The treatment continued for 12 consecutive weeks. Crocin significantly reduced body weight gain and adiposity index as compared to MetS group. Also, crocin protected against the occurrence of hyperglycemia and insulin resistance as indicated by controlled values of HOMA-IR. Crocin protected against MetS-induced dyslipidemia as well as the rise blood pressure. These beneficial effects were accompanied by enhanced serum levels of PPARγ & AMPK and inhibited serum levels of IL-6 and TNF-α. In conclusion, crocin protects against experimentally-induced MetS. This can be attributed, at least partly, to activation of PPARγ and AMPK as well as inhibiting inflammation.
Collapse
Affiliation(s)
- Mardi M Algandaby
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Princess Dr. Najla Bint Saud Al-Saud, Center for Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
17
|
Long Noncoding RNA lnc-HC Regulates PPARγ-Mediated Hepatic Lipid Metabolism through miR-130b-3p. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:954-965. [PMID: 31770672 PMCID: PMC6881655 DOI: 10.1016/j.omtn.2019.10.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/12/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is due to the excessive lipid accumulation within hepatocytes. Metabolic nuclear receptors (MNRs) play great roles in lipid homeostasis. We have identified a novel long noncoding RNA (lncRNA), lnc-HC, which regulates hepatocytic cholesterol metabolism through reducing Cyp7a1 and Abca1 expression. Here, we further elucidate its roles in hepatic fatty acid and triglyceride (TG) metabolism through a novel lncRNA regulatory mechanism. The most prominent target of lnc-HC identified by in vitro study is PPARγ. Further studies revealed that lnc-HC negatively regulates PPARγ at both the mRNA and protein levels and suppresses hepatocytic lipid droplet formation. Importantly, the function of lnc-HC in regulating PPARγ expression depends on modulating miR-130b-3p expression from the transcriptional to the post-transcriptional level, not through lncRNA’s critical modulating patterns. In vivo, the reduction of lnc-HC expression significantly decreases miR-130b-3p expression, induces PPARγ expression, and increases TG concentration in rat livers with hyperlipidemia. These findings further help in understanding the regulatory pattern of lnc-HC in hepatic lipid metabolism and might present a possible therapeutic target for improving lipid homeostasis.
Collapse
|
18
|
Yuan T, Zhang H, Chen D, Chen Y, Lyu Y, Fang L, Du G. Puerarin protects pulmonary arteries from hypoxic injury through the BMPRII and PPARγ signaling pathways in endothelial cells. Pharmacol Rep 2019; 71:855-861. [PMID: 31408784 DOI: 10.1016/j.pharep.2019.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 04/17/2019] [Accepted: 05/06/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Recent evidence indicates that Puerarin has a protective effect on pulmonary arteries. In the present study, we aimed to investigate whether Puerarin could protect pulmonary arterial endothelial cells from hypoxic injury and determine its potential targets. METHODS In our study, human pulmonary arterial endothelial cells (HPAECs) were injured by hypoxic (1% O2) incubation. Cell viability was detected by a cell counting kit (CCK8). The production of nitric oxide (NO) was detected by Griess reagent and endothelin-1 (ET-1) was detected by the ELISA method. Oxidative stress was measured by a fluorescence microscope via the fluorescent probe DCFH-DA. Western blotting was employed for studying the mechanism. RESULTS The results show that Puerarin protects HPAECs from hypoxia-induced apoptosis and slightly improves cell viability. Puerarin increases NO and decreases ET-1 to prevent the imbalance between vasoactive substances induced by hypoxia in HPAECs. Puerarin also inhibits the oxidative stress induced by hypoxia. The results from the Western blot show that Puerarin activates the BMPRII/Smad and PPARγ/PI3K/Akt signaling pathways. CONCLUSION In conclusion, Puerarin protects HPAECs from hypoxic injury through the inhibition of oxidative stress and the activation of the BMPRII and PPARγ signaling pathways. This work provides insight into the development of Puerarin as a treatment for hypoxic pulmonary hypertension.
Collapse
Affiliation(s)
- Tianyi Yuan
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Huifang Zhang
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Di Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Yucai Chen
- Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China
| | - Yang Lyu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Polymorphic Drugs, Beijing, China
| | - Lianhua Fang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China.
| | - Guanhua Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Beijing, China; Beijing Key Laboratory of Drug Targets Identification and Drug Screening, Beijing, China.
| |
Collapse
|
19
|
Han T, Lv Y, Wang S, Hu T, Hong H, Fu Z. PPARγ overexpression regulates cholesterol metabolism in human L02 hepatocytes. J Pharmacol Sci 2019; 139:1-8. [DOI: 10.1016/j.jphs.2018.09.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/17/2018] [Accepted: 09/18/2018] [Indexed: 02/06/2023] Open
|
20
|
Hye Khan MA, Kolb L, Skibba M, Hartmann M, Blöcher R, Proschak E, Imig JD. A novel dual PPAR-γ agonist/sEH inhibitor treats diabetic complications in a rat model of type 2 diabetes. Diabetologia 2018; 61:2235-2246. [PMID: 30032428 PMCID: PMC6563928 DOI: 10.1007/s00125-018-4685-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 06/13/2018] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS The metabolic syndrome is a cluster of risk correlates that can progress to type 2 diabetes. The present study aims to evaluate a novel molecule with a dual action against the metabolic syndrome and type 2 diabetes. METHODS We developed and tested a novel dual modulator, RB394, which acts as a soluble epoxide hydrolase (sEH) inhibitor and a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist in rat models of the metabolic syndrome-the obese spontaneously hypertensive (SHROB) rat and the obese diabetic Zucker fatty/spontaneously hypertensive heart failure F1 hybrid (ZSF1) rat. In SHROB rats we studied the ability of RB394 to prevent metabolic syndrome phenotypes, while in ZSF1 obese diabetic rats we compared RB394 with the ACE inhibitor enalapril in the treatment of type 2 diabetes and associated comorbid conditions. RB394 (10 mg/kg daily) and enalapril (10 mg/kg daily) were administered orally for 8 weeks. RESULTS RB394 blunted the development of hypertension, insulin resistance, hyperlipidaemia and kidney injury in SHROB rats and reduced fasting blood glucose and HbA1c, improved glucose tolerance, reduced blood pressure and improved lipid profiles in obese ZSF1 rats. A reduction in liver fibrosis and hepatosteatosis was evident in RB394-treated obese ZSF1 rats. Unlike RB394, enalapril did not demonstrate any positive effects in relation to diabetes, hyperlipidaemia or liver dysfunction in obese ZSF1 rats. RB394 ameliorated diabetic nephropathy by reducing renal interstitial fibrosis and renal tubular and glomerular injury in obese diabetic ZSF1 rats. Intriguingly, enalapril demonstrated a weaker action against diabetic nephropathy in obese ZSF1 rats. CONCLUSIONS/INTERPRETATION These findings demonstrate that a novel sHE inhibitor/PPAR-γ agonist molecule targets multiple risk factors of the metabolic syndrome and is a glucose-lowering agent with a strong ability to treat diabetic complications.
Collapse
Affiliation(s)
- Md Abdul Hye Khan
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Lauren Kolb
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Melissa Skibba
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Markus Hartmann
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Frankfurt am Main, Germany
| | - René Blöcher
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Frankfurt am Main, Germany
| | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University of Frankfurt, Frankfurt am Main, Germany
| | - John D Imig
- Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
21
|
Construction of lentiviral RNAi vector of PPARγ gene in cashmere goats and comparison with the transcriptome analysis of adipose cells treatments. Small Rumin Res 2018. [DOI: 10.1016/j.smallrumres.2018.04.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
22
|
Interplay between the renin-angiotensin system, the canonical WNT/β-catenin pathway and PPARγ in hypertension. Curr Hypertens Rep 2018; 20:62. [PMID: 29884931 DOI: 10.1007/s11906-018-0860-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Heterogeneous causes can determinate hypertension. RECENT FINDINGS The renin-angiotensin system (RAS) has a major role in the pathophysiology of blood pressure. Angiotensin II and aldosterone are overexpressed during hypertension and lead to hypertension development and its cardiovascular complications. In several tissues, the overactivation of the canonical WNT/β-catenin pathway leads to inactivation of peroxisome proliferator-activated receptor gamma (PPARγ), while PPARγ stimulation induces a decrease of the canonical WNT/β-catenin pathway. In hypertension, the WNT/β-catenin pathway is upregulated, whereas PPARγ is decreased. The WNT/β-catenin pathway and RAS regulate positively each other during hypertension, whereas PPARγ agonists can decrease the expression of both the WNT/β-catenin pathway and RAS. We focus this review on the hypothesis of an opposite interplay between PPARγ and both the canonical WNT/β-catenin pathway and RAS in regulating the molecular mechanism underlying hypertension. The interactions between PPARγ and the canonical WNT/β-catenin pathway through the regulation of the renin-angiotensin system in hypertension may be an interesting way to better understand the actions and the effects of PPARγ agonists as antihypertensive drugs.
Collapse
|
23
|
Ribes-Navarro A, Atef M, Sánchez-Sarasúa S, Beltrán-Bretones MT, Olucha-Bordonau F, Sánchez-Pérez AM. Abscisic Acid Supplementation Rescues High Fat Diet-Induced Alterations in Hippocampal Inflammation and IRSs Expression. Mol Neurobiol 2018; 56:454-464. [PMID: 29721854 DOI: 10.1007/s12035-018-1091-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/17/2018] [Indexed: 01/04/2023]
Abstract
Accumulated evidence indicates that neuroinflammation induces insulin resistance in the brain. Moreover, both processes are intimately linked to neurodegenerative disorders, including Alzheimer's disease. Potential mechanisms underlying insulin resistance include serine phosphorylation of the insulin receptor substrate (IRS) or insulin receptor (IR) misallocation. However, only a few studies have focused on IRS expression in the brain and its modulation in neuroinflammatory processes. This study used the high-fat diet (HFD) model of neuroinflammation to study the alterations of IR, an insulin-like growth factor receptor (IGF1R) and IRS expressions in the hippocampus. We observed that HFD effectively reduced mRNA and protein IRS2 expression. In contrast, a HFD induced the upregulation of the IRS1 mRNA levels, but did not alter an IR and IGF1R expression. As expected, we observed that a HFD increased hippocampal tumor necrosis factor alpha (TNFα) and amyloid precursor protein (APP) levels while reducing brain-derived neurotrophic factor (BDNF) expression and neurogenesis. Interestingly, we found that TNFα correlated positively with IRS1 and negatively with IRS2, whereas APP levels correlated positively only with IRS1 but not IRS2. These results indicate that IRS1 and IRS2 hippocampal expression can be affected differently by HFD-induced neuroinflammation. In addition, we aimed to establish whether abscisic acid (ABA) can rescue hippocampal IRS1 and IRS2 expression, as we had previously shown that ABA supplementation prevents memory impairments and improves neuroinflammation induced by a HFD. In this study, ABA restored HFD-induced hippocampal alterations, including IRS1 and IRS2 expression, TNFα, APP, and BDNF levels and neurogenesis. In conclusion, this study highlights different regulations of hippocampal IRS1 and IRS2 expression using a HFD, indicating the important differences of these scaffolding proteins, and strongly supports ABA therapeutic effects.
Collapse
Affiliation(s)
| | - Mariam Atef
- Department of Medicine, University of Jaume I, Castellón de la Plana, Spain
| | | | | | | | | |
Collapse
|
24
|
Shen Q, Li J, Zhang C, Wang P, Mohammed A, Ni S, Tang Z. Panax notoginseng saponins reduce high-risk factors for thrombosis through peroxisome proliferator-activated receptor -γ pathway. Biomed Pharmacother 2017; 96:1163-1169. [PMID: 29174034 DOI: 10.1016/j.biopha.2017.11.106] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 11/01/2017] [Accepted: 11/20/2017] [Indexed: 12/09/2022] Open
Abstract
The classic Virchow theory suggests that blood stasis, hypercoagulability and endothelial dysfunction are three major factors that cause venous thrombosis (VT). It is a complicated biological process involved multi-factors. Platelet plays a central role and participates in multiple links of this process. Panax notoginseng saponins (PNS), the principal constituents derived from panax notoginseng, has been widely described for its anti-platelet activity. However, its potential mechanism against platelet aggregation has not been clarified. In this present study, we evaluated the anti-platelet effects of PNS on thrombin-induced platelet activation and its possible molecular mechanism of action, and further explored the therapeutic action of PNS on thrombin induced hypercoagulability in rat. Our results showed that PNS treatment inhibited platelet aggregation induced by thrombin, which was accompanied with over-expression of Peroxisome proliferator-activated receptor γ (PPAR-γ) protein, mRNA and upregulation of phosphatidylinositol 3 kinase (PI3K)/ protein kinase B (Akt)/ endothelial nitric oxide synthase (eNOS) pathway in platelet, and this effect could be reversed by PPAR-γ inhibitor T0070907. In vivo, PNS significantly reversed thrombin-induced hypercoagulable state in rat which was accompanied by PPAR-γ protein and mRNA upregulation in rat lung. In conclusion, these data suggested that PNS could suppress thrombin-induced platelet aggregation in vitro and effectively improve hypercoagulable state in vivo and PNS-induced activation of PPAR-γ and its downstream PI3K/Akt/eNOS pathway played the central role.
Collapse
Affiliation(s)
- Qin Shen
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jun Li
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Caixin Zhang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Pengbo Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Anaz Mohammed
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Songshi Ni
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| | - Zhiyuan Tang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
25
|
Magvanjav O, Gong Y, McDonough CW, Chapman AB, Turner ST, Gums JG, Bailey KR, Boerwinkle E, Beitelshees AL, Tanaka T, Kubo M, Pepine CJ, Cooper-DeHoff RM, Johnson JA. Genetic Variants Associated With Uncontrolled Blood Pressure on Thiazide Diuretic/β-Blocker Combination Therapy in the PEAR (Pharmacogenomic Evaluation of Antihypertensive Responses) and INVEST (International Verapamil-SR Trandolapril Study) Trials. J Am Heart Assoc 2017; 6:e006522. [PMID: 29097388 PMCID: PMC5721751 DOI: 10.1161/jaha.117.006522] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/11/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND The majority of hypertensive individuals require combination antihypertensive therapy to achieve adequate blood pressure (BP) control. This study aimed to identify genetic variants associated with uncontrolled BP on combination therapy with a thiazide diuretic and a β-blocker. METHODS AND RESULTS A genome-wide association study of uncontrolled BP on combination therapy was conducted among 314 white participants of the PEAR (Pharmacogenomic Evaluation of Antihypertensive Responses) trial. Multivariable logistic regression analysis was used. Genetic variants meeting a suggestive level of significance (P<1.0E-05) were tested for replication in an external cohort, INVEST (International Verapamil-SR Trandolapril study). We also examined genome-wide variant associations with systolic and diastolic BP response on combination therapy and tested for replication. We discovered a single nucleotide polymorphism, the rs261316 major allele, at chromosome 15 in the gene ALDH1A2 associated with an increased odds of having uncontrolled BP on combination therapy (odds ratio: 2.56, 95% confidence interval, 1.69-3.88, P=8.64E-06). This single nucleotide polymorphism replicated (odds ratio: 1.86, 95% confidence interval, 1.35-2.57, P=0.001) and approached genome-wide significance in the meta-analysis between discovery and replication cohorts (odds ratio: 2.16, 95% confidence interval, 1.63-2.86, P=8.60E-08). Other genes in the region surrounding rs261316 (ALDH1A2) include AQP9 and LIPC. CONCLUSIONS A single nucleotide polymorphism in the gene ALDH1A2 may be associated with uncontrolled BP following treatment with a thiazide diuretic/β-blocker combination. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov. Unique identifier: NCT00246519.
Collapse
Affiliation(s)
- Oyunbileg Magvanjav
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL
- College of Medicine, University of Florida, Gainesville, FL
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL
| | - Caitrin W McDonough
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL
| | - Arlene B Chapman
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, IL
| | - Stephen T Turner
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN
| | - John G Gums
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL
| | - Kent R Bailey
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Eric Boerwinkle
- Human Genetics Center and Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX
| | - Amber L Beitelshees
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland School of Medicine, Baltimore, MD
| | | | - Michiaki Kubo
- RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Carl J Pepine
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Rhonda M Cooper-DeHoff
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL
| | - Julie A Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics, University of Florida College of Pharmacy, Gainesville, FL
- Division of Cardiovascular Medicine, Department of Medicine, University of Florida College of Medicine, Gainesville, FL
| |
Collapse
|
26
|
Becer E, Çırakoğlu A. Effect of the Pro12Ala Polymorphism of the Peroxisome Proliferator-activated Receptor γ2 Gene on Lipid Profile and Adipokines Levels in Obese Subjects. Balkan J Med Genet 2017; 20:71-80. [PMID: 28924543 PMCID: PMC5596824 DOI: 10.1515/bjmg-2017-0007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a key regulator of metabolism, adipokines production and secretion. The aim of this study was to investigate the association between the PPARγ2 gene Pro12Ala polymorphism in obesity in terms of body mass index (BMI), lipid parameters, homeostasis model assessment of insulin resistance (HOMA-IR), serum lipid, leptin, adiponectin, resistin and chemerin levels. The study included 160 obese and 140 non obese subjects. The Pro12Ala polymorphism was determined by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP). Serum lipid, leptin, adiponectin, resistin and chemerin levels were measured. No association was found between the Pro12Ala polymorphism and BMI. Strikingly, in the study group, obese subjects with the AA genotype had significantly higher triglycerides (p = 0.046) and resistin (p <0.001) levels than those with the wild-type PP and heterozygous PA genotypes. Serum leptin and chemerin levels were significantly associated with Pro-12Ala poymorphism in the obese and non obese groups (p <0.01). In the obese group, subjects with the homozygous AA genotype had significantly lower adiponectin (p = 0.010) activity than the PP genotype. Our results suggest that the PPARγ2 gene Pro12Ala polymorphism has no direct association with obesity but does have significant influences on lipid profiles and adipokines levels.
Collapse
Affiliation(s)
- E Becer
- Department of Biochemistry, Faculty of Pharmacy, Near East University, Nicosia, Mersin 10, Turkey.,Experimental Health Science Research Center, Near East University, Nicosia, Mersin 10, Turkey
| | - A Çırakoğlu
- Department of Medical Biology, Cerrahpasa Faculty of Medicine,Istanbul University, Istanbul, Turkey
| |
Collapse
|
27
|
Wang J, Ke W, Bao R, Hu X, Chen F. Beneficial effects of ginger Zingiber officinale Roscoe on obesity and metabolic syndrome: a review. Ann N Y Acad Sci 2017; 1398:83-98. [PMID: 28505392 DOI: 10.1111/nyas.13375] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/07/2017] [Accepted: 04/12/2017] [Indexed: 01/16/2023]
Abstract
In recent years, metabolic syndromes (MetSs), including diabetes mellitus, dyslipidemia, and cardiovascular diseases, have become a common health problem in both developed and developing countries. Accumulating data have suggested that traditional herbs might be able to provide a wide range of remedies in prevention and treatment of MetSs. Ginger (Zingiber officinale Roscoe, Zingiberaceae) has been documented to ameliorate hyperlipidemia, hyperglycemia, oxidative stress, and inflammation. These beneficial effects are mediated by transcription factors, such as peroxisome proliferator-activated receptors, adenosine monophosphate-activated protein kinase, and nuclear factor κB. This review focuses on recent findings regarding the beneficial effects of ginger on obesity and related complications in MetS and discusses its potential mechanisms of action. This review provides guidance for further applications of ginger for personalized nutrition and medicine.
Collapse
Affiliation(s)
- Jing Wang
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing, Ministry of Agriculture; and Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing, China
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Weixin Ke
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing, Ministry of Agriculture; and Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Rui Bao
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing, Ministry of Agriculture; and Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing, China
| | - Xiaosong Hu
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing, Ministry of Agriculture; and Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing, China
- College of Food Science and Nutritional Engineering, Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Fang Chen
- College of Food Science and Nutritional Engineering, National Engineering Research Center for Fruit and Vegetable Processing, Key Laboratory of Fruit and Vegetable Processing, Ministry of Agriculture; and Engineering Research Centre for Fruit and Vegetable Processing, Ministry of Education, China Agricultural University, Beijing, China
| |
Collapse
|
28
|
Holzer G, Markov GV, Laudet V. Evolution of Nuclear Receptors and Ligand Signaling. Curr Top Dev Biol 2017; 125:1-38. [DOI: 10.1016/bs.ctdb.2017.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
29
|
Atypical Rho GTPases of the RhoBTB Subfamily: Roles in Vesicle Trafficking and Tumorigenesis. Cells 2016; 5:cells5020028. [PMID: 27314390 PMCID: PMC4931677 DOI: 10.3390/cells5020028] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/07/2016] [Accepted: 06/08/2016] [Indexed: 12/12/2022] Open
Abstract
RhoBTB proteins constitute a subfamily of atypical Rho GTPases represented in mammals by RhoBTB1, RhoBTB2, and RhoBTB3. Their characteristic feature is a carboxyl terminal extension that harbors two BTB domains capable of assembling cullin 3-dependent ubiquitin ligase complexes. The expression of all three RHOBTB genes has been found reduced or abolished in a variety of tumors. They are considered tumor suppressor genes and recent studies have strengthened their implication in tumorigenesis through regulation of the cell cycle and apoptosis. RhoBTB3 is also involved in retrograde transport from endosomes to the Golgi apparatus. One aspect that makes RhoBTB proteins atypical among the Rho GTPases is their proposed mechanism of activation. No specific guanine nucleotide exchange factors or GTPase activating proteins are known. Instead, RhoBTB might be activated through interaction with other proteins that relieve their auto-inhibited conformation and inactivated through auto-ubiquitination and destruction in the proteasome. In this review we discuss our current knowledge on the molecular mechanisms of action of RhoBTB proteins and the implications for tumorigenesis and other pathologic conditions.
Collapse
|
30
|
Abstract
The central nervous system (CNS) in concert with the heart and vasculature is essential to maintaining cardiovascular (CV) homeostasis. In recent years, our understanding of CNS control of blood pressure regulation (and dysregulation leading to hypertension) has evolved substantially to include (i) the actions of signaling molecules that are not classically viewed as CV signaling molecules, some of which exert effects at CNS targets in a non-traditional manner, and (ii) CNS locations not traditionally viewed as central autonomic cardiovascular centers. This review summarizes recent work implicating immune signals and reproductive hormones, as well as gasotransmitters and reactive oxygen species in the pathogenesis of hypertension at traditional CV control centers. Additionally, recent work implicating non-conventional CNS structures in CV regulation is discussed.
Collapse
Affiliation(s)
- Pauline M Smith
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L3N6, Canada
| | - Alastair V Ferguson
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, K7L3N6, Canada
| |
Collapse
|