1
|
Li Z, Wu Y, Qian M, Zhang B, Deng X, Mao P, Fan Z, Fang X, Cheng L, Liu X, Wang L, Liu H. Multi-omics analysis reveals BPF exposure causes hepatic glucose and lipid metabolism disorder in rats by disrupting energy homeostasis. Toxicology 2025; 515:154130. [PMID: 40188933 DOI: 10.1016/j.tox.2025.154130] [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: 01/23/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/12/2025]
Abstract
Bisphenol F (BPF) is one of the main substitutes for Bisphenol A (BPA) and is widely used in the manufacture of household products. In addition, BPF threatens human health through environmental pollution and the food chain. However, the hepatotoxicity of BPF and its effects on glucose and lipid metabolism remain unclear. This study used male SD rats as an animal model to investigate the hepatotoxicity of BPF and its effects on glucose and lipid metabolism. The results of the HE staining, serum and liver biochemical indicators show that BPF can damage the basic structure of the liver, cause liver dysfunction and lead to disorders of liver glucose metabolism and lipid metabolism. Furthermore, we conducted metabolomics and proteomics analyses on the livers of the BPF exposed group at 100 mg/kg/d in comparison with the control group. The results indicated that BPF exposure had a significant effect on liver metabolism. Combined with biological analysis and the validation of changes in genes and proteins related to glucose and lipid metabolism in the liver, it was elucidated that BPF can promote fatty acid oxidation and inhibit fatty acid synthesis through the AMPK and PPAR signaling pathways, leading to a reduction in fatty acids. Furthermore, it has been demonstrated that BPF can promote glycogen synthesis and gluconeogenesis via the AKT pathway, which can result in disorders of glucose metabolism.
Collapse
Affiliation(s)
- Zhi Li
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Yuanyuan Wu
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Mingqing Qian
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Bingya Zhang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China
| | - Xinxin Deng
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Penghui Mao
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Zhonghua Fan
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China
| | - Xu Fang
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Lin Cheng
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Xuan Liu
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China
| | - Li Wang
- School of Public Health, Bengbu Medical University, Bengbu 233030, PR China.
| | - Hui Liu
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical University, Bengbu 233030, PR China; Bengbu Medical University Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu 233030, PR China.
| |
Collapse
|
2
|
Feng S, Tang J, Wei X, Lu Z, Xu Y, Zhang T, Han H. Swertia cincta and its main active ingredients regulate the PPAR-α pathway in anti-cholestatic liver injury. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118956. [PMID: 39423946 DOI: 10.1016/j.jep.2024.118956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Swertia cincta is a traditional remedy for cholestasis commonly utilised in Yunnan, China. Despite its widespread use, the specific active components and underlying mechanisms of action remain poorly understood. AIM OF THIS STUDY This study aimed to investigate the therapeutic properties, mechanisms, and active compounds of Swertia cincta in an animal model of cholestasis induced by alpha-naphthylisothiocyanate (ANIT). MATERIALS AND METHODS UHPLC/Q-TOF-MS and high-performance liquid chromatography (HPLC) were utilised to analyse the blood components of Swertia cincta. An ANIT-induced cholestatic liver injury animal model was established, and metabolomics was employed to explore the potential mechanisms of Swertia cincta in treating cholestatic liver injury. Hepatocellular injury induced by taurochenodeoxycholic acid was evaluated in vitro, and key bioactive components of Swertia cincta for cholestatic liver injury treatment were identified and confirmed using the ANIT-induced mouse model. RESULTS The established HPLC method demonstrates good specificity and reproducibility, enabling the simultaneous determination of six components in Swertia cincta. Results from serum biochemical indicators and liver pathology analysis indicated that Swertia cincta exhibits promising anti-cholestasis liver injury effects. Specifically, gentiopicroside, loganic acid, and isoorientin were identified as key active ingredients in treating cholestatic liver injury. Their mechanism of action primarily involves regulating PPAR-α, FXR, CYP3A4, NTCP, CAR, and CPT2. By modulating PPAR-α and bile acid metabolism-related proteins, reducing pro-inflammatory factors, enhancing bile acid transport, and promoting fatty acid oxidation to reduce lipid accumulation, Swertia cincta exerts protective and therapeutic effects against cholestatic liver injury. Notably, gentian bitter glycosides appear to be the most critical components for this effect. CONCLUSION Swertia cincta may improve cholestatic liver injury by activating the peroxisome proliferator-activated receptor alpha pathway, and the key active compounds were gentiopicroside, loganic acid, and isoorientin.
Collapse
Affiliation(s)
- Shuaixia Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Jie Tang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Xia Wei
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Zou Lu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Ying Xu
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| | - Han Han
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
| |
Collapse
|
3
|
Dai FF, Chen J, Ma Z, Yang MH, Sun T, Ma J, Zhou MJ, Wei ZR, Zou Y, Zhang S, Zang MX. The polycomb protein complex interacts with GATA-6/PPARα to inhibit α-MHC expression. Dev Growth Differ 2025; 67:23-32. [PMID: 39723530 DOI: 10.1111/dgd.12953] [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: 04/09/2024] [Revised: 11/27/2024] [Accepted: 12/07/2024] [Indexed: 12/28/2024]
Abstract
Transcription factors collaborate with epigenetic regulatory factors to orchestrate cardiac differentiation for heart development, but the underlying mechanism is not fully understood. Here, we report that GATA-6 induces cardiac differentiation but peroxisome proliferator-activated receptor α (PPARα) reverses GATA-6-induced cardiac differentiation, possibly because GATA-6/PPARα recruits the polycomb protein complex containing EZH2/Ring1b/BMI1 to the promoter of the cardiac-specific α-myosin heavy chain (α-MHC) gene and suppresses α-MHC expression, which ultimately inhibits cardiac differentiation. Furthermore, Ring1b ubiquitylates PPARα and GATA-6. By overexpression and knockout of EZH2/BMI1, it was demonstrated that the polycomb protein complex inhibits cardiac differentiation induced by GATA-6 and PPARα. Together, our results demonstrate that the polycomb protein complex interacts with GATA-6/PPARα to inhibit cardiac differentiation, a finding that could facilitate the development of new therapies for congenital heart disease.
Collapse
Affiliation(s)
- Fei-Fei Dai
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Chen
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhen Ma
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Ming-Hui Yang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Tong Sun
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Juan Ma
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Meng-Jiao Zhou
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhi-Ru Wei
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shoutao Zhang
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Ming-Xi Zang
- Department of Biochemistry & Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Birth Defects Prevention, Zhengzhou, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Fudan University, Shanghai, China
| |
Collapse
|
4
|
Zhu J, Wang L, Guo Z, Zhang T, Zhang P. Transcriptome analysis of intestine from alk-SMase knockout mice reveals the effect of alk-SMase. Cancer Cell Int 2022; 22:344. [DOI: 10.1186/s12935-022-02764-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 10/11/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Objective
Intestinal alkaline sphingomyelinase (alk-SMase) generates ceramide and inactivates platelet-activating factor associated with digestion and inhibition of cancer. There is few study to analyze the correlated function and characterize the genes related to alk-SMase comprehensively. We characterised transcriptome landscapes of intestine tissues from alk-SMase knockout (KO) mice aiming to identify novel associated genes and research targets.
Methods
We performed the high-resolution RNA sequencing of alk-SMase KO mice and compared them to wild type (WT) mice. Differentially expressed genes (DEGs) for the training group were screened. Functional enrichment analysis of the DEGs between KO mice and WT mice was implemented using the Database for Annotation, Visualization and Integrated Discovery (DAVID). An integrated protein–protein interaction (PPI) and Kyoto Encyclopedia of Genes and Genomes (KEGG) network was chose to study the relationship of differentially expressed gene. Moreover, quantitative real-time polymerase chain reaction (qPCR) was further used to validate the accuracy of RNA-seq technology.
Results
Our RNA-seq data found 97 differentially expressed mRNAs between the WT mice and alk-SMase gene NPP7 KO mice, in which 32 were significantly up-regulated and 65 were down-regulated, including protein coding genes, non-coding RNAs. Notably, the results of gene ontology functional enrichment analysis indicated that DEGs were functionally associated with the immune response, regulation of cell proliferation and development related terms. Additionally, an integrated network analysis was shown that some modules was significantly related to alk-SMase and with accordance of previously results. We chose 6 of these genes randomly were validated the accuracy of RNA-seq technology using qPCR and 2 genes showed difference significantly (P < 0.05).
Conclusions
We investigated the potential biological significant of alk-SMase with high resolution genome-wide transcriptome of alk-SMase knockout mice. The results revealed new insight into the functional modules related to alk-SMase was involved in the intestinal related diseases.
Collapse
|
5
|
Zhu J, Wang L, Li X, Lan D, Song L, Li Y, Cheng Y, Zhang P. Transcriptome analysis alk-SMase knockout mice reveals the effect of alkaline sphingomyelinase on liver. Biochem Biophys Rep 2022; 30:101240. [PMID: 35360085 PMCID: PMC8961189 DOI: 10.1016/j.bbrep.2022.101240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/21/2022] [Accepted: 02/26/2022] [Indexed: 11/25/2022] Open
|
6
|
Cizkova K, Koubova K, Tauber Z. Lipid Messenger Phosphatidylinositol-4,5-Bisphosphate Is Increased by Both PPARα Activators and Inhibitors: Relevance for Intestinal Cell Differentiation. BIOLOGY 2022; 11:biology11070997. [PMID: 36101378 PMCID: PMC9312331 DOI: 10.3390/biology11070997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 11/16/2022]
Abstract
Simple Summary Fibrates, such as fenofibrate, are widely used drugs for dyslipidaemia treatment. It is known that they activate peroxisome proliferator-activated receptor α (PPARα) which serves as a lipid sensor in the organism. This article addresses how activators and inhibitor of the PPARα could affect differentiation of intestinal cells. Carcinogenesis is a disruption of normal differentiation process and colorectal carcinoma is the third most common cancer in terms of incidence, but the secondp in terms of mortality. One of the important signalling pathways in intestinal cell differentiation as well as carcinogenesis is PI3K/Akt/PTEN. We showed that PPARα activators as well as inhibitor affected the levels of one member of this pathway called phosphatidylinositol-4,5-bisphosphate. This molecule is important for formation of microvilli, the essential structures of fully differentiated intestinal cells. Abstract We investigated the effects of PPARα activators fenofibrate and WY-14643 as well as the PPARα inhibitor GW6471 on the PI3K/Akt/PTEN pathway of intestinal cell differentiation. Our previous study showed that all these compounds increased the expression of villin, a specific marker of intestinal cell differentiation in HT-29 and Caco2 cells. Our current results confirmed the central role of lipid messenger phosphatidylinositol-4,5-bisphosphate (PIP2), a known player in brush border formation, in mediating the effects of tested PPARα ligands. Although all tested compounds increased its levels, surprisingly, each of them affected different PIP2-metabolizing enzymes, especially the levels of PIP5K1C and PTEN. Moreover, we found a positive relationship between the expression of PPARα itself and PIP2 as well as PIP5K1C. By contrast, PPARα was negatively correlated with PTEN. However, the expression of antigens of interest was independent of PPARα subcellular localization, suggesting that it is not directly involved in their regulation. In colorectal carcinoma tissues we found a decrease in PTEN expression, which was accompanied by a change in its subcellular localization. This change was also observed for the regulatory subunit of PI3K. Taken together, our data revealed that fenofibrate, WY-14643, and GW6471 affected different members of the PI3K/Akt/PTEN pathway. However, these effects were PPARα-independent.
Collapse
|
7
|
Cizkova K, Foltynkova T, Hanyk J, Kamencak Z, Tauber Z. When Activator and Inhibitor of PPARα Do the Same: Consequence for Differentiation of Human Intestinal Cells. Biomedicines 2021; 9:biomedicines9091255. [PMID: 34572440 PMCID: PMC8472525 DOI: 10.3390/biomedicines9091255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/13/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Peroxisome proliferator-activated receptor α (PPARα) is a ligand-dependent transcription factor that plays a role in various processes including differentiation of several cell types. We investigated the role of PPARα in the differentiation of intestinal cells using HT-29 and Caco2 cell lines as a model as well as human normal colon and colorectal carcinoma tissues. We detected a significant increase in PPARα expression in differentiated HT-29 cells as well as in normal surface colon epithelium where differentiated cells are localised. Thus, it seems that PPARα may play a role in differentiation of intestinal cells. Interestingly, we found that both PPARα activators (fenofibrate and WY-14643) as well as its inhibitor (GW6471) regulated proliferation and differentiation of HT-29 cells in vitro in the same way. Both compounds led to a decrease in proliferation accompanied by a significant increase in expression of villin, intestinal alkaline phosphatase (differentiation markers). Moreover, the same trend in villin expression was observed in Caco2 cells. Furthermore, villin expression was independent of subcellular localisation of PPARα. In addition, we found similar levels of PPARα expression in colorectal carcinomas in comparison to adjacent normal epithelium. All these findings support the hypothesis that differentiation of intestinal epithelium is PPARα-independent.
Collapse
Affiliation(s)
| | | | | | | | - Zdenek Tauber
- Correspondence: ; Tel.: +420-585-632-283; Fax: +420-585-632-966
| |
Collapse
|
8
|
Zhang W, Li JY, Wei XC, Wang Q, Yang JY, Hou H, Du ZW, Wu XA. Effects of dibutyl phthalate on lipid metabolism in liver and hepatocytes based on PPARα/SREBP-1c/FAS/GPAT/AMPK signal pathway. Food Chem Toxicol 2021; 149:112029. [PMID: 33508418 DOI: 10.1016/j.fct.2021.112029] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 01/02/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023]
Abstract
Phateacid esters (PAEs), such as dibutyl phthalate (DBP), have been widely used and human exposure results into serious toxic effects; such as the development of fatty liver disease. In the present study, SD rat models for in vivo study (normal and fatty liver model group) and hepatocytes for in vitro study (normal and abnormal lipid metabolism model group) were established to determine the effects of DBP on liver function and discover the possible mechanisms. Meanwhile, the peroxisome proliferator activated receptor (PPARα) blocker, GW6471, with the Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) activator, AICAR, were applied in vitro study to clarify the role of PPARα/SREBP-1c/FAS/GPAT/AMPK signal pathway in the process. Results suggested that DBP could activate PPARα signaling pathway and affected the protein expression of SREBP, FAS and GPAT to cause hyperlipidemia and abnormal liver function. DBP also could inhibit the phosphorylation and activation of AMPK to inhibit the decomposition and metabolism of lipids. Interestingly, the effects of DBP could be alleviated by GW6471 and AICAR. Our experimental results provide reliable evidence that DBP exposure could further induce liver lipid metabolism disorder and other hepatic toxicity through PPARα/SREBP-1c/FAS/GPAT/AMPK signal pathway.
Collapse
Affiliation(s)
- Wang Zhang
- Department of Pharmacy, Hefei BOE Hospital, Hefei, PR China
| | - Jing-Ya Li
- Institute of Intelligent Machines, Chinese Academy of Sciences, Hefei, 230031, China; Department of Biological Physics, University of Science and Technology of China, Hefei, 230026, China
| | - Xiao-Chen Wei
- Department of Pharmacy, Hefei BOE Hospital, Hefei, PR China
| | - Qian Wang
- Department of Pharmacy, Hefei BOE Hospital, Hefei, PR China
| | - Ji-Yang Yang
- Department of Pharmacy, Hefei BOE Hospital, Hefei, PR China
| | - Huan Hou
- Department of Pharmacy, Hefei BOE Hospital, Hefei, PR China
| | - Zi-Wei Du
- Department of Pharmacy, Hefei BOE Hospital, Hefei, PR China
| | - Xin-An Wu
- Department of Pharmacy, Hefei BOE Hospital, Hefei, PR China.
| |
Collapse
|
9
|
Malandraki-Miller S, Lopez CA, Alonaizan R, Purnama U, Perbellini F, Pakzad K, Carr CA. Metabolic flux analyses to assess the differentiation of adult cardiac progenitors after fatty acid supplementation. Stem Cell Res 2019; 38:101458. [PMID: 31102832 PMCID: PMC6618003 DOI: 10.1016/j.scr.2019.101458] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 12/13/2022] Open
Abstract
Myocardial infarction is the most prevalent of cardiovascular diseases and pharmacological interventions do not lead to restoration of the lost cardiomyocytes. Despite extensive stem cell therapy studies, clinical trials using cardiac progenitor cells have shown moderate results. Furthermore, differentiation of endogenous progenitors to mature cardiomyocytes is rarely reported. A metabolic switch from glucose to fatty acid oxidation occurs during cardiac development and cardiomyocyte maturation, however in vitro differentiation protocols do not consider the lack of fatty acids in cell culture media. The aim of this study was to assess the effect of this metabolic switch on control and differentiated adult cardiac progenitors, by fatty acid supplementation. Addition of oleic acid stimulated the peroxisome proliferator-activated receptor alpha pathway and led to maturation of the cardiac progenitors, both before and after transforming growth factor-beta 1 differentiation. Addition of oleic acid following differentiation increased expression of myosin heavy chain 7 and connexin 43. Also, total glycolytic metabolism increased, as did mitochondrial membrane potential and glucose and fatty acid transporter expression. This work provides new insights into the importance of fatty acids, and of peroxisome proliferator-activated receptor alpha, in cardiac progenitor differentiation. Harnessing the oxidative metabolic switch induced maturation of differentiated endogenous stem cells. (200 words).
Collapse
Affiliation(s)
- Sophia Malandraki-Miller
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Colleen A Lopez
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Rita Alonaizan
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Ujang Purnama
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Filippo Perbellini
- National Heart and Lung Institute, Imperial College London, London, W12 0NN, UK.
| | - Kathy Pakzad
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| | - Carolyn A Carr
- Department of Physiology, Anatomy, and Genetics,Sherrington Building, University of Oxford, Oxford, UK.
| |
Collapse
|
10
|
Wang Z, Shi H, Zhao H, Dong Z, Zhao B, Weng X, Liu R, Hu K, Zou Y, Sun A, Ge J. Naoxintong Retards Atherosclerosis by Inhibiting Foam Cell Formation Through Activating Pparα Pathway. Curr Mol Med 2018; 18:698-710. [PMID: 30734676 PMCID: PMC6463403 DOI: 10.2174/1566524019666190207143207] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 01/08/2019] [Accepted: 01/24/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUNDS We recently reported that Naoxintong (NXT), a China Food and Drug Administration (FDA)-approved cardiac medicine, could reduce the plaque size, but the underlying mechanism remains elusive now. OBJECTIVE In this study, we investigated the effects of NXT on foam cell accumulation both in vivo and in vitro and explored related mechanisms. METHOD THP-1 cells and bone marrow-derived macrophages were incubated with oxidized low-density lipoprotein (ox-LDL) with/without Naoxintong. ApoE-/- mice fed an atherogenic diet were administered to receive NXT for eight weeks. Macrophage-derived foam cell formation in plaques was measured by immunohistochemical staining. Expression of proteins was evaluated by Western blot. Lentivirus was used to knockdown PPARα in THP-1 cells. RESULTS After NXT treatment, foam cell accumulation was significantly reduced in atherosclerotic plaques. Further investigation revealed that oxidized low-density lipoprotein (ox-LDL) uptake was significantly decreased and expression of scavenger receptor class A (SR-A) and class B (SR-B and CD36) was significantly downregulated post-NXT treatment. On the other hand, NXT increased cholesterol efflux and upregulated ATP-binding cassette (ABC) transporters (ABCA-1 and ABCG-1) in macrophages. Above beneficial effects of NXT were partly abolished after lentiviral knockdown of PPARα. CONCLUSION Our findings suggest that NXT could retard atherosclerosis by inhibiting foam cell formation through reducing ox-LDL uptake and enhancing cholesterol efflux and above beneficial effects are partly mediated through PPARα pathway.
Collapse
Affiliation(s)
- Zeng Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
| | - Huairui Shi
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
| | - Huan Zhao
- Department of Pathology, LiShui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical College, ZheJiang, China
| | - Zhen Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
| | - Buchang Zhao
- Shandong Buchang Pharmaceutical Co., Ltd, Shandong, China
| | - Xinyu Weng
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Rongle Liu
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
| | - Xiao lia
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
- Department of Pathology, LiShui Central Hospital, The Fifth Affiliated Hospital of Wenzhou Medical College, ZheJiang, China
- Shandong Buchang Pharmaceutical Co., Ltd, Shandong, China
| | - Kai Hu
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
| | - Aijun Sun
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
| | - Junbo Ge
- Institutes of Biomedical Sciences, Fudan University, Shanghai200032, China
- Department of Cardiology, Zhongshan Hospital, Fudan University. Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai200032, P.R. China
| |
Collapse
|
11
|
Peymani M, Ghaedi K, Irani S, Nasr-Esfahani MH. Peroxisome Proliferator-Activated Receptor γ Activity is Required for Appropriate Cardiomyocyte Differentiation. CELL JOURNAL 2016; 18:221-8. [PMID: 27540527 PMCID: PMC4988421 DOI: 10.22074/cellj.2016.4317] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Accepted: 10/22/2015] [Indexed: 11/16/2022]
Abstract
Objective Peroxisome proliferator-activated receptor γ (PPARγ) is a member of the
PPAR nuclear receptor superfamily. Although PPARγ acts as a master transcription factor
in adipocyte differentiation, it is also associated with a variety of cell functions including
carbohydrate and lipid metabolism, glucose homeostasis, cell proliferation and cell differentiation. This study aimed to assess the expression level of PPARγ in order to address its
role in cardiac cell differentiation of mouse embryonic stem cells (mESCs).
Materials and Methods In this an intervening study, mESCs were subjected to cardiac differentiation. Total RNA was extracted from the cells and quantitative real time polymerase chain
reaction (qPCR) was carried out to estimate level of gene expression. Furthermore, the requirement of PPARγ in cardiac differentiation of mESCs, during cardiac progenitor cells (CPCs)
formation, was examined by applying the respective agonist and antagonist.
Results The obtained data revealed an elevation in the expression level of PPARγ during
spontaneous formation of CPCs and cardiomyocytes. Our results indicated that during
CPC formation, PPARγ inactivation via treatment with GW9662 (GW) reduced expression
of CPC and cardiac markers.
Conclusion We conclude that PPARγ modulation has an effective role on cardiac differentiation of mESCs at the early stage of cardiomyogenesis.
Collapse
Affiliation(s)
- Maryam Peymani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran; Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
12
|
Shabani P, Ghazizadeh Z, Pahlavan S, Hashemizadeh S, Baharvand H, Aghdami N, Doosti M. Exogenous treatment with eicosapentaenoic acid supports maturation of cardiomyocytes derived from embryonic stem cells. Biochem Biophys Res Commun 2015; 461:281-6. [PMID: 25871791 DOI: 10.1016/j.bbrc.2015.04.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/03/2015] [Indexed: 11/29/2022]
Abstract
Embryonic stem cells offer multiple advantages over adult stem cells in terms of achieving acceptable number of functional cardiomyocytes to be exploited in cell therapy. However, differentiation efficacy is still a major issue to be solved before moving to regenerative medicine. Although a vast number of chemical compounds have been tested on efficiency of cardiac differentiation, the effect of fish oil components, such as eicosapentaenoic acid (EPA) on developmental bioenergetics, and hence cardiac differentiation, remained unstudied. EPA has been reported to have several cardioprotective effects, but there is no study addressing its role in cardiac differentiation. After mesoderm induction of embryoid bodies (EBs) derived from mouse embryonic stem cells (mESCs) in hanging drops initiated by ascorbic acid, they were treated with various concentrations of EPA. Gene and protein expression and functional properties of cardiomyocytes derived from ESCs were evaluated following treatment with various concentrations of EPA. Exposure to low concentrations of EPA (10 μM) increased percentage of beating colonies and beating area. This treatment also resulted in up to 3 fold increase in expression of NKX2-5, MEF2C, MYH6, TNNT2 and CX43. FACS analysis confirmed gene expression analysis with increased percentage of MYH6 positive cells in EPA-treated group compared to the control group. In contrast, the expression of genes coding for cardiac differentiation, remained constant or even declined with higher concentrations of EPA. In conclusion, we have demonstrated that treatment of mESCs undergoing cardiac differentiation with low concentration, but not high concentration of EPA up-regulate transcription of genes associated with cardiac development.
Collapse
Affiliation(s)
- Parisa Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran; Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Zaniar Ghazizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shiva Hashemizadeh
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nasser Aghdami
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Mahmood Doosti
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
13
|
Liu L, Ma Y, Wang RL, Xu WR, Wang SQ, Chou KC. Find novel dual-agonist drugs for treating type 2 diabetes by means of cheminformatics. Drug Des Devel Ther 2013; 7:279-88. [PMID: 23630413 PMCID: PMC3623550 DOI: 10.2147/dddt.s42113] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The high prevalence of type 2 diabetes mellitus in the world as well as the increasing reports about the adverse side effects of the existing diabetes treatment drugs have made developing new and effective drugs against the disease a very high priority. In this study, we report ten novel compounds found by targeting peroxisome proliferator-activated receptors (PPARs) using virtual screening and core hopping approaches. PPARs have drawn increasing attention for developing novel drugs to treat diabetes due to their unique functions in regulating glucose, lipid, and cholesterol metabolism. The reported compounds are featured with dual functions, and hence belong to the category of dual agonists. Compared with the single PPAR agonists, the dual PPAR agonists, formed by combining the lipid benefit of PPARα agonists (such as fibrates) and the glycemic advantages of the PPARγ agonists (such as thiazolidinediones), are much more powerful in treating diabetes because they can enhance metabolic effects while minimizing the side effects. This was observed in the studies on molecular dynamics simulations, as well as on absorption, distribution, metabolism, and excretion, that these novel dual agonists not only possessed the same function as ragaglitazar (an investigational drug developed by Novo Nordisk for treating type 2 diabetes) did in activating PPARα and PPARγ, but they also had more favorable conformation for binding to the two receptors. Moreover, the residues involved in forming the binding pockets of PPARα and PPARγ among the top ten compounds are explicitly presented, and this will be very useful for the in-depth conduction of mutagenesis experiments. It is anticipated that the ten compounds may become potential drug candidates, or at the very least, the findings reported here may stimulate new strategies or provide useful insights for designing new and more powerful dual-agonist drugs for treating type 2 diabetes.
Collapse
Affiliation(s)
- Lei Liu
- PET/CT Center, General Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | | | | | | | | | | |
Collapse
|
14
|
AlSuleimani YM, Hiley CR. Mechanisms of vasorelaxation induced by oleoylethanolamide in the rat small mesenteric artery. Eur J Pharmacol 2013; 702:1-11. [DOI: 10.1016/j.ejphar.2013.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 12/20/2012] [Accepted: 01/09/2013] [Indexed: 10/27/2022]
|
15
|
Liang X, Mei Y, Huang X, Shen G, Zhu D, Yu Y, Wang J, Lou Y. Junctophilin 2 knockdown interfere with mitochondrium status in ESC-CMs and cardiogenesis of ES cells. J Cell Biochem 2012; 113:2884-94. [DOI: 10.1002/jcb.24164] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
16
|
Rose BA, Force T, Wang Y. Mitogen-activated protein kinase signaling in the heart: angels versus demons in a heart-breaking tale. Physiol Rev 2010; 90:1507-46. [PMID: 20959622 PMCID: PMC3808831 DOI: 10.1152/physrev.00054.2009] [Citation(s) in RCA: 563] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Among the myriad of intracellular signaling networks that govern the cardiac development and pathogenesis, mitogen-activated protein kinases (MAPKs) are prominent players that have been the focus of extensive investigations in the past decades. The four best characterized MAPK subfamilies, ERK1/2, JNK, p38, and ERK5, are the targets of pharmacological and genetic manipulations to uncover their roles in cardiac development, function, and diseases. However, information reported in the literature from these efforts has not yet resulted in a clear view about the roles of specific MAPK pathways in heart. Rather, controversies from contradictive results have led to a perception that MAPKs are ambiguous characters in heart with both protective and detrimental effects. The primary object of this review is to provide a comprehensive overview of the current progress, in an effort to highlight the areas where consensus is established verses the ones where controversy remains. MAPKs in cardiac development, cardiac hypertrophy, ischemia/reperfusion injury, and pathological remodeling are the main focuses of this review as these represent the most critical issues for evaluating MAPKs as viable targets of therapeutic development. The studies presented in this review will help to reveal the major challenges in the field and the limitations of current approaches and point to a critical need in future studies to gain better understanding of the fundamental mechanisms of MAPK function and regulation in the heart.
Collapse
Affiliation(s)
- Beth A Rose
- Departments of Anesthesiology, Physiology, and Medicine, David Geffen School of Medicine, Molecular Biology, Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | |
Collapse
|