1
|
Saidi S, Remok F, Handaq N, Drioiche A, Gourich AA, Menyiy NE, Amalich S, Elouardi M, Touijer H, Bouhrim M, Bouissane L, Nafidi HA, Bin Jardan YA, Bourhia M, Zair T. Phytochemical Profile, Antioxidant, Antimicrobial, and Antidiabetic Activities of Ajuga iva (L.). Life (Basel) 2023; 13:life13051165. [PMID: 37240812 DOI: 10.3390/life13051165] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/03/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
In Morocco, many applications in ethnomedicine on Ajuga iva (L.) have been recognized as able to treat various pathologies such as diabetes, stress, and microbial infections. The objective of this work is to carry out phytochemical, biological, and pharmacological investigations on the extracts of Ajuga iva leaves in order to confirm its therapeutic effects. The phytochemical screening carried out on the different extracts of Ajuga iva showed its richness in primary (lipids and proteins) and secondary metabolites (flavonoids, tannins, reducing compounds, oses, and holoside. The best contents of polyphenols, flavonoids, and tannins evaluated by spectrophotometric methods were found in the hydroethanolic extract (69.850 ± 2.783 mg EAG/g DE, 17.127 ± 0.474 mg EQ/g DE, 5.566 ± 0.000 mg EQC/g DE), respectively. Analysis of the chemical composition of the aqueous extract by LC/UV/MS revealed 32 polyphenolic compounds including ferulic acid (19.06%), quercetin (10.19%), coumaric acid (9.63%), and apigenin-7-(2-O-apiosylglucoside) (6.8%). The antioxidant activity of Ajuga iva extracts was evaluated by three methods (DPPH*, FRAP, CAT). The hydroethanolic extract recorded the strongest reducing power: DPPH* (IC50 = 59.92 ± 0.7 µg/mL), FRAP (EC50 = 196.85 ± 1.54 (µg/mL), and CAT (199.21 ± 0.37 mg EAG/gE). A strong correlation between phenolic compounds and antioxidant activities was confirmed by the determination of Pearson's coefficient. The antimicrobial activity of Ajuga iva studied by the microtiter method revealed potent antifungal and antibacterial qualities against Candida parapsilosis and Staphylococcus aureus BLACT. An in vivo oral glucose tolerance test (OGTT) using normal rats revealed that the antihyperglycemic action of the aqueous extract significantly reduced postprandial hyperglycaemia at (30 min, p < 0.01) and area under the curve (AUC glucose), p < 0.01. Similarly, the aqueous extract, tested on pancreatic α-amylase enzyme activity in vitro and in vivo significantly inhibited pancreatic α-amylase activity with IC50 = 1.52 ± 0.03 mg/mL. In conclusion, the extract from Ajuga iva could be a good source of bioactive molecules, which exhibit potent antioxidant and antimicrobial activity, as well as strong antidiabetic activity, for applications in the pharmaceutical industry.
Collapse
Affiliation(s)
- Soukaina Saidi
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
- Laboratory of Molecular Chemistry, Materials and Catalysis, Faculty of Science and Technologies, Sultan Moulay Slimane University, Beni Mellal 23000, Morocco
| | - Firdaous Remok
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
| | - Nadia Handaq
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
- Plant Valorization and Protection Research Team, Laboratory of Environmental Biology and Sustainable Development, Higher Normal School of Tetouan, Abdelmaek Essaadi University, Tetouan 93000, Morocco
| | - Aziz Drioiche
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
| | - Aman Allah Gourich
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
| | - Naoual El Menyiy
- Laboratory of Pharmacology and Phytochemistry, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco
| | - Smail Amalich
- Laboratory of Pharmacology and Phytochemistry, National Agency of Medicinal and Aromatic Plants, Taounate 34025, Morocco
| | - Mohamed Elouardi
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
| | - Hanane Touijer
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
| | - Mohamed Bouhrim
- Laboratory of Biological Engineering, Team of Functional and Pathological Biology, Faculty of Sciences and Technology Beni Mellal, University Sultan Moulay Slimane, Beni Mellal 23000, Morocco
| | - Latifa Bouissane
- Laboratory of Molecular Chemistry, Materials and Catalysis, Faculty of Science and Technologies, Sultan Moulay Slimane University, Beni Mellal 23000, Morocco
| | - Hiba-Allah Nafidi
- Department of Food Science, Faculty of Agricultural and Food Sciences, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Yousef A Bin Jardan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11495, Saudi Arabia
| | - Mohammed Bourhia
- Department of Chemistry and Biochemistry, Faculty of Medicine and Pharmacy, Laayoune 70000, Morocco
| | - Touriya Zair
- Research Team of Bioactive Molecules and Environment Chemistry, Laboratory of Innovative Materials and Biotechnology of Natural Resources, Faculty of Sciences, Moulay Ismail University, Meknes 50070, Morocco
| |
Collapse
|
2
|
Jung F, Braune S, Jung CHG, Krüger-Genge A, Waldeck P, Petrick I, Küpper JH. Lipophilic and Hydrophilic Compounds from Arthrospira platensis and Its Effects on Tissue and Blood Cells-An Overview. Life (Basel) 2022; 12:1497. [PMID: 36294932 PMCID: PMC9605678 DOI: 10.3390/life12101497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 09/14/2024] Open
Abstract
The cyanobacterium Arthrospira platensis (Spirulina platensis) is a natural source of considerable amounts of ingredients that are relevant for nutra- and pharmaceutical uses. Different hydrophilic and hydrophobic substances can be obtained by extraction from the biomass. The respective extraction techniques determine the composition of substances in the extract and thus its biological activity. In this short review, we provide an overview of the hydrophilic compounds (phenols, phycobiliproteins, polysaccharides, and vitamins) and lipophilic ingredients (chlorophylls, vitamins, fatty acids, and glycolipids) of Arthrospira platensis. The principal influences of these substances on blood and tissue cells are briefly summarized.
Collapse
Affiliation(s)
- Friedrich Jung
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Steffen Braune
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | | | - Anne Krüger-Genge
- Department of Healthcare, Biomaterials and Cosmeceuticals, Fraunhofer-Institute for Applied Polymer Research, 14476 Potsdam-Golm, Germany
| | - Peter Waldeck
- Institute of Materials Chemistry, Thermodynamics, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Ingolf Petrick
- Institute of Materials Chemistry, Thermodynamics, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
| | - Jan-Heiner Küpper
- Institute of Biotechnology, Molecular Cell Biology, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- Faculty of Health Sciences Brandenburg, Brandenburg University of Technology Cottbus-Senftenberg, 01968 Senftenberg, Germany
- Carbon Biotech Social Enterprise AG, 01968 Senftenberg, Germany
| |
Collapse
|
3
|
Jin X, Zhen Z, Wang Z, Gao X, Li M. GPRC6A is a key mediator of palmitic acid regulation of lipid synthesis in bovine mammary epithelial cells. Cell Biol Int 2022; 46:1747-1758. [PMID: 35979663 DOI: 10.1002/cbin.11886] [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: 10/02/2021] [Revised: 12/29/2021] [Accepted: 03/07/2022] [Indexed: 11/07/2022]
Abstract
Fatty acids (FAs) can promote lipid synthesis in the mammary gland via stimulating lipogenic gene expression, but the underlying molecular mechanism is still not fully understood. Here, we showed the dose-dependent effects of palmitic acid (PA) on lipid synthesis in primary bovine mammary epithelial cells (BMECs) and explored the corresponding molecular mechanism. BMECs were treated with PA (0, 50, 100, 150, and 200 μM), and the 100 μM treatment had the best stimulatory effect on lipid synthesis and expression and maturation of sterol regulatory element-binding protein 1c (SREBP-1c) in cells. Inhibition of phosphatidylinositol 3-kinase (PI3K) almost totally blocked the stimulation of PA on SREBP-1c expression, whereas protein kinase Cα (PKCα) knockdown only partially decreased the stimulation of PA on SREBP-1c expression but abolished the stimulation of PA on its maturation. Knockdown of GPR120 did not change the stimulation of PA on the SREBP-1c signaling. G protein-coupled receptor family C group 6 member A (GPRC6A) knockdown almost totally blocked the stimulation of FA on PI3K and PKCα phosphorylation as well as SREBP-1c expression and maturation. Furthermore, PA dose-dependently promoted GPRC6A expression and plasma membrane localization. Together, these above results reveal that GPRC6A is a key mediator of PA signaling to lipid synthesis in BMECs via the PI3K/PKCα-SREBP-1c pathways.
Collapse
Affiliation(s)
- Xin Jin
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhen Zhen
- College of Life Science, Northeast Agricultural University, Harbin, China
| | - Zhaoxiong Wang
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Xuejun Gao
- College of Animal Science, Yangtze University, Jingzhou, China
| | - Meng Li
- College of Life Science, Northeast Agricultural University, Harbin, China
| |
Collapse
|
4
|
Xu X, Luo D, Xuan Q, Lu P, Yu C, Guan Q. Atlas of metabolism reveals palmitic acid results in mitochondrial dysfunction and cell apoptosis by inhibiting fatty acid β-oxidation in Sertoli cells. Front Endocrinol (Lausanne) 2022; 13:1021263. [PMID: 36237186 PMCID: PMC9552013 DOI: 10.3389/fendo.2022.1021263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
In recent years, the impact of lipotoxicity on male fertility has received extensive attention, especially on Sertoli cells (SCs). In SCs, energy metabolism is important as disorders of energy metabolism result in infertility eventually. However, the underlying mechanism of lipotoxicity on energy metabolism in SCs remains unknown. Advances in high-throughput metabolomics and lipidomics measurement platforms provide powerful tools to gain insights into complex biological systems. Here, we aimed to explore the potential molecular mechanisms of palmitic acid (PA) regulating energy metabolism in SCs based on metabolomics and lipidomics. The results showed that glucose metabolism-related metabolites were not significantly changed, which suggested that PA treatment had little effect on glucose metabolism and may not influence the normal energy supply from SCs to germ cells. However, fatty acid β-oxidation was inhibited according to accumulation of medium- and long-chain acylcarnitines in cells. In addition, the pool of amino acids and the levels of most individual amino acids involved in the tricarboxylic acid (TCA) cycle were not changed after PA treatment in SCs. Moreover, PA treatment of SCs significantly altered the lipidome, including significant decreases in cardiolipin and glycolipids as well as remarkable increases in ceramide and lysophospholipids, which indicated that mitochondrial function was affected and apoptosis was triggered. The increased apoptosis rate of SCs was verified by elevated protein expression levels of Cleaved Caspase-3 and Bax as well as decreased Bcl-2 protein expression level. Together, these findings indicated that PA may result in mitochondrial dysfunction and increased apoptosis by inhibiting fatty acid β-oxidation of SCs.
Collapse
Affiliation(s)
- Xiaoqin Xu
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
| | - Dandan Luo
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiuhui Xuan
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Peng Lu
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Chunxiao Yu
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Qingbo Guan, ; Chunxiao Yu,
| | - Qingbo Guan
- Shandong Provincial Hospital, Shandong University, Jinan, China
- Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Laboratory of Endocrinology and Lipid Metabolism, Shandong Provincial Hospital, Jinan, China
- Shandong Prevention and Control Engineering Laboratory of Endocrine and Metabolic Diseases, Shandong Provincial Hospital, Jinan, China
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Qingbo Guan, ; Chunxiao Yu,
| |
Collapse
|
5
|
Overexpression CPT1A reduces lipid accumulation via PPARα/CD36 axis to suppress the cell proliferation in ccRCC. Acta Biochim Biophys Sin (Shanghai) 2021; 54:220-231. [PMID: 35130611 PMCID: PMC9909300 DOI: 10.3724/abbs.2021023] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Clear cell renal carcinoma (ccRCC) is histologically defined by its cytoplasmic lipid deposits. Lipid metabolism disorder largely increases the risk of ccRCC. In this study, we aimed to investigate the biological functions and molecular mechanisms of carnitine palmitoyl transferase 1A (CPT1A) in ccRCC. Our results showed that CPT1A is decreased in ccRCC clinical samples and cell lines compared with that in normal samples. Lentivirus overexpressing CPT1A was used to investigate the neoplastic phenotypes of ccRCC, and the results showed that lipid accumulation and tumor growth are attenuated both and . In addition, CPT1A prevents cholesterol uptake and lipid accumulation by increasing the peroxisome proliferator-activated receptor α (PPARα) level through regulation of Class B scavenger receptor type 1 (SRB1) and cluster of differentiation 36 (CD36). Furthermore, PI3K/Akt signaling pathway promotes tumor cell proliferation in ccRCC, which is related to the enhanced expression of CD36. Functionally, weakened CPT1A expression is critical for lipid accumulation to promote ccRCC development. Collectively, our research unveiled a novel function of CPT1A in lipid metabolism via PPARα/CD36 axis, which provides a new theoretical explanation for the pathogenesis of ccRCC. Targeting CPT1A may be a potential therapeutic strategy to treat ccRCC.
Collapse
|
6
|
Moroccan antidiabetic medicinal plants: Ethnobotanical studies, phytochemical bioactive compounds, preclinical investigations, toxicological validations and clinical evidences; challenges, guidance and perspectives for future management of diabetes worldwide. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.03.032] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
7
|
The Anti-Inflammatory Effect of Zhibaidihuang Decoction on Recurrent Oral Ulcer with Sirt1 as the Key Regulatory Target. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8886699. [PMID: 34007301 PMCID: PMC8110403 DOI: 10.1155/2021/8886699] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 04/10/2021] [Accepted: 04/19/2021] [Indexed: 01/07/2023]
Abstract
The syndrome of ROU is generally manifested as obvious pain, redness, and swelling of local ulceration area, accompanied by flushed face, red eyes, sore throat, and swollen gums. Traditional Chinese medicine (TCM) doctors believe that "yin deficiency" is one causative factor of ROU. Zhibaidihuang decoction (ZBDHD) is a prescriptively developed receipt, where Anemarrhena asphodeloides and Phellodendri amurensis Cortex are added in the original Liuweidihuang decoction. It is generally used for "yin deficiency" treatment. It can effectively reduce the recurrence of oral ulcers and release the severity of the disease. However, the mechanism of this activity remains to be elucidated. In this study, we found that ZBDHD has a certain therapeutic effect on the pathological changes of oral mucosa. Furthermore, the results of serum metabolomics showed ZBDHD influenced the synthesis and metabolism of certain fatty acids. The results of western blot, immunochemical, and immunofluorescence staining indicate that ZBDHD could increase the expression of Sirt1 and Foxp3 and suppress the expression and acetylation of NF-κB in oral mucosa cells. By screening active ingredients in ZBDHD, we found berberine, as well as other compounds, presenting high fitness of the Sirt1 reactive centre. Therefore, it is possible that ZBDHD can regulate the Sirt1-NF-κB pathway to improve fatty acids metabolism in the body, thereby achieving the effect of treating ROU.
Collapse
|
8
|
Sabu V, Krishnan S, Peter J, Aswathy IS, Lal Preethi SS, Simon M, Radhakrishna GP, Helen A. Synergistic effect of Betulinic acid, Apigenin and Skimmianine (BASk) in high cholesterol diet rabbit: Involvement of CD36-TLR2 signaling pathway. Cytokine 2021; 142:155475. [PMID: 33667961 DOI: 10.1016/j.cyto.2021.155475] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 02/05/2021] [Accepted: 02/13/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Progression of chronic inflammatory disease, atherosclerosis is a multifactorial process. Cluster of differentiation 36 (CD36) mediated downstream activation of Toll like receptor 2 (TLR2) and NLRP3 (Nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3) inflammasome signaling pathway actively participates during chronic inflammation. Nowadays, synergistic combinations of bioactive compounds attained priority in the field of drug discovery and development as therapeutic agents. An investigation regarding the anti-inflammatory potential of a novel drug formulation, BASk which is a combination of three bioactive compounds Betulinic acid (B):Apigenin (A):Skimmianine (Sk) remains the focus area of this research study. We also elucidate the molecular mechanism behind the therapeutic potential of BASk through CD36 mediated activation TLR2-NLRP3 signaling pathway. METHODS OxLDL induced hPBMCs used to screen out a suitable combination of BASk via MTT, COX, LOX, NOS and MPO assays. Hypercholesterolemia is induced in rabbits by supplementing with 1% cholesterol + 0.5% cholic acid and treated with BASk (2:2:1) (5 mg/Kg) and atorvastatin (10 mg/Kg) for 60 days. CD36, TLR2, NLRP3, NFκB, cytokines, endothelial damage were quantified by reverse transcription, real time PCR, ELISA, flow cytometry and histopathology. RESULTS hPBMCs pretreated with BASk at 2:2:1 ratio significantly decreased the activities of COX, 15-LOX, NOS and MPO on OxLDL induction than quercetin. Down regulation of CD36, TLR2, MyD88, TRAF6 by BASk further buttressed NLRP3 inflammasome activation mediated by the transcription factor NFκB. This is in correlation with the effect of BASk by balancing pro (IL-1β, IL-18) and anti-inflammatory (TGF-β) mediators in the aortic endothelial cells. CONCLUSION BASk exerted its anti-inflammatory potential by reducing pro-inflammatory mediators during cholesterol supplementation via down regulating CD36 mediated TLR2 - NLRP3 inflammasome cascade. This deciphers a synergistic combination named BASk (2:2:1) as a novel drug formulation against chronic inflammatory disease, atherosclerosis.
Collapse
Affiliation(s)
- Vidya Sabu
- Department of Biochemistry, Research Centre, University of Kerala, Kariavattom Campus, Thiruvananthapuram, 695581 Kerala, India
| | - Santhi Krishnan
- Department of Biochemistry, Research Centre, University of Kerala, Kariavattom Campus, Thiruvananthapuram, 695581 Kerala, India
| | - Jasmine Peter
- Department of Biochemistry, Research Centre, University of Kerala, Kariavattom Campus, Thiruvananthapuram, 695581 Kerala, India
| | - I S Aswathy
- Department of Biochemistry, Research Centre, University of Kerala, Kariavattom Campus, Thiruvananthapuram, 695581 Kerala, India
| | - S S Lal Preethi
- Department of Biochemistry, Research Centre, University of Kerala, Kariavattom Campus, Thiruvananthapuram, 695581 Kerala, India
| | - Monisha Simon
- Department of Biochemistry, Research Centre, University of Kerala, Kariavattom Campus, Thiruvananthapuram, 695581 Kerala, India
| | | | - A Helen
- Department of Biochemistry, Research Centre, University of Kerala, Kariavattom Campus, Thiruvananthapuram, 695581 Kerala, India.
| |
Collapse
|
9
|
Metabolomics coupled with integrated approaches reveal the therapeutic effects of higenamine combined with [6]-gingerol on doxorubicin-induced chronic heart failure in rats. Chin Med 2020; 15:120. [PMID: 33292391 PMCID: PMC7670783 DOI: 10.1186/s13020-020-00403-0] [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: 05/25/2020] [Accepted: 11/11/2020] [Indexed: 12/19/2022] Open
Abstract
Background This study was aimed to investigate the therapeutic effects and potential mechanism of higenamine combined with [6]-gingerol (HG/[6]-GR) against doxorubicin (DOX)—induced chronic heart failure (CHF) in rats. Materials and methods Therapeutic effects of HG/[6]-GR on hemodynamics indices, serum biochemical indicators, histopathology and TUNEL staining of rats were assessed. Moreover, a UHPLC-Q-TOF/MS-based serum metabolic approach was performed to identify the metabolites and possible pathways of HG/[6]-GR on DOX-induced CHF. Results HG/[6]-GR had effects on regulating hemodynamic indices, alleviating serum biochemical indicators, improving the pathological characteristics of heart tissue and reducing the apoptosis of myocardial cells. Serum metabolisms analyses indicated that the therapeutic effects of HG and [6]-GR were mainly associated with the regulation of eight metabolites, including acetylphosphate, 3-Carboxy-1-hydroxypropylthiamine diphosphate, coenzyme A, palmitic acid, PE(O-18:1(1Z)/20:4(5Z,8Z,11Z,14Z)), oleic acid, lysoPC(18:1(9Z)), and PC(16:0/16:0). Pathway analysis showed that HG/[6]-GR on CHF treatment was related to twelve pathways, including glycerophospholipid metabolism, fatty acid metabolism, pantothenate and CoA biosynthesis, citrate cycle (TCA cycle), pyruvate metabolism, and arachidonic acid metabolism. Serum metabolites and metabolic pathways regulated by HG/[6]-GR appear to be related to energy metabolism. Conclusion Multivariate statistical analysis has provided new insights for understanding CHF and investigating the therapeutic effects and mechanisms of HG/[6]-GR, which influencing the metabolites and pathways related to energy metabolism pathway.
Collapse
|
10
|
Cardiac Transcriptome Analysis Reveals Nr4a1 Mediated Glucose Metabolism Dysregulation in Response to High-Fat Diet. Genes (Basel) 2020; 11:genes11070720. [PMID: 32610475 PMCID: PMC7397175 DOI: 10.3390/genes11070720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/23/2022] Open
Abstract
Obesity is associated with an increased risk of developing cardiovascular disease (CVD), with limited alterations in cardiac genomic characteristics known. Cardiac transcriptome analysis was conducted to profile gene signatures in high-fat diet (HFD)-induced obese mice. A total of 184 differentially expressed genes (DEGs) were identified between groups. Based on the gene ontology (GO) term enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis of DEGs, the critical role of closely interlocked glucose metabolism was determined in HFD-induced cardiac remodeling DEGs, including Nr4a1, Fgf21, Slc2a3, Pck1, Gck, Hmgcs2, and Bpgm. Subsequently, the expression levels of these DEGs were evaluated in both the myocardium and palmitic acid (PA)-stimulated H9c2 cardiomyocytes using qPCR. Nr4a1 was highlighted according to its overexpression resulting from the HFD. Additionally, inhibition of Nr4a1 by siRNA reversed the PA-induced altered expression of glucose metabolism-related DEGs and hexokinase 2 (HK2), the rate-limiting enzyme in glycolysis, thus indicating that Nr4a1 could modulate glucose metabolism homeostasis by regulating the expression of key enzymes in glycolysis, which may subsequently influence cardiac function in obesity. Overall, we provide a comprehensive understanding of the myocardium transcript molecular framework influenced by HFD and propose Nr4a1 as a key glucose metabolism target in obesity-induced CVD.
Collapse
|
11
|
Wu RM, Jiang B, Li H, Dang WZ, Bao WL, Li HD, Ye G, Shen X. A network pharmacology approach to discover action mechanisms of Yangxinshi Tablet for improving energy metabolism in chronic ischemic heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2020; 246:112227. [PMID: 31509780 DOI: 10.1016/j.jep.2019.112227] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 09/07/2019] [Accepted: 09/08/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Most cardiovascular diseases ultimately result in heart failure, an intractable problem in modern medicine. Yangxinshi tablet (YXS) is a Chinese medicine formula that is used clinically to treat coronary heart disease. However, the active compounds, potential targets, and pharmacological and molecular mechanism of its anti-heart failure activity remain unclear. Therefore, further investigation is required. AIM OF STUDY Active ingredients and potential targets of YXS for treating heart failure have been reported previously. However, the molecular functions or biological processes of YXS in energy metabolism have not been discovered. To date, no experimental study to validate the potential anti-heart failure mechanism of YXS. The aim of this study was to study the therapeutic effect of YXS on rats with chronic ischemic heart failure by evaluating rat cardiac function and exercise tolerance, and to explore its potential mechanism by network pharmacology, western blotting, quantitative RT-PCR and histological analysis. MATERIALS AND METHODS In this investigation, chronic ischemic heart failure rats were randomly assigned to five groups: control group (sham operation), model group (0.5% CMC-Na), trimetazidine group (positive control) and two YXS groups (low- and high-dose groups). Experimental rats were treated by gavage with 10 mg/kg/d (clinical equivalent dose) trimetazidine (TMZ), 500 mg/kg/d (clinical equivalent dose) YXS and 1000 mg/kg/d YXS, respectively, for 5 weeks. The cardiac functions of rats were detected by High-Resolution In Vivo Imaging System. We elucidated novel understanding of the active compounds of YXS in rat plasma and predicted the energy metabolism related targets and processes for heart failure. Then, we validated experimentally the targets and mechanism of YXS on these pathological processes in vivo. RESULTS It was found that YXS was able to effectively improve cardiac LVIDs, LVEDV, LVESV and EF, decrease myocardial oxygen consumption and reduce myocardial infarct size in rats with chronic ischemic heart failure was similar to that of TMZ. We identified 63 major candidate targets for YXS that are closely to heart failure progression. Enrichment analysis revealed key targets for YXS associated to oxygen delivery, glucose utilization, and mitochondrial biogenesis. Meanwhile, we validated that YXS could promote the expression of downstream HIF-1α, PGC1α and GLUT4 by increasing phosphorylation of PI3K, Akt, mTOR, rpS6 and AMPK. The results show that YXS could activate related PI3K/Akt/mTOR/rpS6/HIF-1α and AMPK/PGC1α/GLUT4 signaling pathways in chronic ischemic heart failure rats. Further experiments demonstrated that YXS increased mitochondrial biogenesis in chronic ischemic heart failure rats and improved exercise tolerance CONCLUSION: YXS treated chronic ischemic heart failure through activating its targets which play pivotal roles in oxygen delivery, glucose utilization and mitochondrial biogenesis to improve energy metabolism through a multi-component, multi-level, multi-target, multi-pathway and multi-mechanism approaches.
Collapse
Affiliation(s)
- Ruo-Ming Wu
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd, Shanghai, China
| | - Bing Jiang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China; Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hui Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wen-Zhen Dang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Wei-Lian Bao
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Hai-Dong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Guan Ye
- Central Research Institute, Shanghai Pharmaceuticals Holding Co., Ltd, Shanghai, China
| | - Xiaoyan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.
| |
Collapse
|
12
|
Xia P, Cao K, Hu X, Liu L, Yu D, Dong S, Du J, Xu Y, Liu B, Yang Y, Gao F, Sun X, Liu H. K ATP Channel Blocker Glibenclamide Prevents Radiation-Induced Lung Injury and Inhibits Radiation-Induced Apoptosis of Vascular Endothelial Cells by Increased Ca 2+ Influx and Subsequent PKC Activation. Radiat Res 2019; 193:171-185. [PMID: 31877256 DOI: 10.1667/rr15381.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Radiation-induced lung injury (RILI) is a common and severe side effect of thoracic radiotherapy, which compromises patients' quality of life. Recent studies revealed that early vascular injury, especially microvascular damage, played a central role in the development of RILI. For this reason, early vascular protection is essential for RILI therapy. The ATP-sensitive K+ (KATP) channel is an ATP-dependent K+ channel with multiple subunits. The protective role of the KATP channel in vascular injury has been demonstrated in some published studies. In this work, we investigated the effect of KATP channel on RILI. Our findings confirmed that the KATP channel blocker glibenclamide, rather than the KATP channel opener pinacidil, remitted RILI, and in particular, provided protection against radiation-induced vascular injury. Cytology experiments verified that glibenclamide enhanced cell viability, increased the potential of proliferation after irradiation and attenuated radiation-induced apoptosis. Involved mechanisms included increased Ca2+ influx and PKC activation, which were induced by glibenclamide pretreatment. In conclusion, the KATP channel blocker glibenclamide remitted RILI and inhibited the radiation-induced apoptosis of vascular endothelial cells by increased Ca2+ influx and subsequent PKC activation.
Collapse
Affiliation(s)
- Penglin Xia
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Kun Cao
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xuguang Hu
- Department of Gastrointestinal Surgery, Changhai Hospital, Shanghai, P.R. China
| | - Lei Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Danyang Yu
- Ophthalmology Department of Kunming General Hospital of Chengdu Military Area Command, Kunming, Yunnan, China
| | - Suhe Dong
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Jicong Du
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Yang Xu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Bin Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Yanyong Yang
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Fu Gao
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xuejun Sun
- Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Hu Liu
- Department of Radiation Medicine, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| |
Collapse
|
13
|
Mellal K, Omri S, Mulumba M, Tahiri H, Fortin C, Dorion MF, Pham H, Garcia Ramos Y, Zhang J, Pundir S, Joyal JS, Bouchard JF, Sennlaub F, Febbraio M, Hardy P, Gravel SP, Marleau S, Lubell WD, Chemtob S, Ong H. Immunometabolic modulation of retinal inflammation by CD36 ligand. Sci Rep 2019; 9:12903. [PMID: 31501473 PMCID: PMC6733801 DOI: 10.1038/s41598-019-49472-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 08/20/2019] [Indexed: 12/26/2022] Open
Abstract
In subretinal inflammation, activated mononuclear phagocytes (MP) play a key role in the progression of retinopathies. Little is known about the mechanism involved in the loss of photoreceptors leading to vision impairment. Studying retinal damage induced by photo-oxidative stress, we observed that cluster of differentiation 36 (CD36)-deficient mice featured less subretinal MP accumulation and attenuated photoreceptor degeneration. Moreover, treatment with a CD36-selective azapeptide ligand (MPE-001) reduced subretinal activated MP accumulation in wild type mice and preserved photoreceptor layers and function as assessed by electroretinography in a CD36-dependent manner. The azapeptide modulated the transcriptome of subretinal activated MP by reducing pro-inflammatory markers. In isolated MP, MPE-001 induced dissociation of the CD36-Toll-like receptor 2 (TLR2) oligomeric complex, decreasing nuclear factor-kappa B (NF-κB) and NLR family pyrin domain containing 3 (NLRP3) inflammasome activation. In addition, MPE-001 caused an aerobic metabolic shift in activated MP, involving peroxisome proliferator-activated receptor-γ (PPAR-γ) activation, which in turn mitigated inflammation. Accordingly, PPAR-γ inhibition blocked the cytoprotective effect of MPE-001 on photoreceptor apoptosis elicited by activated MP. By altering activated MP metabolism, MPE-001 decreased immune responses to alleviate subsequent inflammation-dependent neuronal injury characteristic of various vision-threatening retinal disorders.
Collapse
Affiliation(s)
- Katia Mellal
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | - Samy Omri
- Maisonneuve-Rosemont Hospital, Montréal, Canada
- Mperia Therapeutics, Montréal, Canada
| | | | - Houda Tahiri
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | - Carl Fortin
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | | | - Hung Pham
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | | | - Jinqiang Zhang
- Department of Chemistry, Université de Montréal, Montreal, Canada
| | - Sheetal Pundir
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | - Jean-Sébastien Joyal
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | - Jean-François Bouchard
- Neuropharmacology Laboratory, School of Optometry, Université de Montréal, Montreal, Canada
| | - Florian Sennlaub
- Institut de la Vision, Sorbonne Universités, INSERM, CNRS, Paris, France
| | - Maria Febbraio
- Department of Dentistry, University of Alberta, Edmonton, Canada
| | - Pierre Hardy
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada
| | | | - Sylvie Marleau
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montreal, Canada
| | - Sylvain Chemtob
- Maisonneuve-Rosemont Hospital, Montréal, Canada.
- Departments of Pediatrics, Ophthalmology and Pharmacology, Université de Montréal, Montreal, Canada.
| | - Huy Ong
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada.
| |
Collapse
|
14
|
Nifedipine Modulates Renal Lipogenesis via the AMPK-SREBP Transcriptional Pathway. Int J Mol Sci 2019; 20:ijms20071570. [PMID: 30934807 PMCID: PMC6480582 DOI: 10.3390/ijms20071570] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/16/2019] [Accepted: 03/27/2019] [Indexed: 12/13/2022] Open
Abstract
Lipid accumulation in renal cells has been implicated in the pathogenesis of obesity-related kidney disease, and lipotoxicity in the kidney can be a surrogate marker for renal failure or renal fibrosis. Fatty acid oxidation provides energy to renal tubular cells. Ca2+ is required for mitochondrial ATP production and to decrease reactive oxygen species (ROS). However, how nifedipine (a calcium channel blocker) affects lipogenesis is unknown. We utilized rat NRK52E cells pre-treated with varying concentrations of nifedipine to examine the activity of lipogenesis enzymes and lipotoxicity. A positive control exposed to oleic acid was used for comparison. Nifedipine was found to activate acetyl Coenzyme A (CoA) synthetase, acetyl CoA carboxylase, long chain fatty acyl CoA elongase, ATP-citrate lyase, and 3-hydroxy-3-methyl-glutaryl-coenzyme A (HMG CoA) reductase, suggesting elevated production of cholesterol and phospholipids. Nifedipine exposure induced a vast accumulation of cytosolic free fatty acids (FFA) and stimulated the production of reactive oxygen species, upregulated CD36 and KIM-1 (kidney injury molecule-1) expression, inhibited p-AMPK activity, and triggered the expression of SREBP-1/2 and lipin-1, underscoring the potential of nifedipine to induce lipotoxicity with renal damage. To our knowledge, this is the first report demonstrating nifedipine-induced lipid accumulation in the kidney.
Collapse
|
15
|
Yao Y, Liu T, Wang X, Zhang D. The Contrary Effects of Sirt1 on MCF7 Cells Depend on CD36 Expression Level. J Surg Res 2019; 238:248-254. [PMID: 30826489 DOI: 10.1016/j.jss.2019.01.046] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 11/16/2018] [Accepted: 01/17/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Breast cancer is one of the most aggressive and pervasive cancers identified in females. Sirt1 and CD36 both exert an essential role toward the oncogenic signaling in breast cancer cells. As reported, the adrenergic signaling could promote the malignancy of breast cancer. This study focuses specifically on the role of Sirt1/CD36 in the proliferation of MCF-7 breast cancer cells and also investigates their response to the α2-adrenergic agonist dexmedetomidine (Dex). MATERIALS AND METHODS Expression of Sirt1 and CD36 was measured in breast cancer tissue by immunohistochemistry. We cultured MCF7 cells and treated cells with resveratrol (RSV) or Dex. Western blot analysis was performed to quantify the protein expression levels. The methyl thiazolyl tetrazolium (MTT) assay was applied to detect cell proliferation. RESULTS Compared with normal adjacent tissues, Sirt1 increased and CD36 decreased in cancer tissues. RSV, a Sirt1 activator, increased the proliferation of MCF-7 cells at low concentration but exerted cytotoxicity effect at higher concentration. Sirt1 activation increased the expression of CD36 at higher concentration. Dex treatment gradually increased the proliferation of MCF7 cells in a dose-dependent manner and downregulated the expression of Sirt1/CD36. Interestingly, overexpression of Sirt1 via RSV pretreatment could suppress Dex-stimulated proliferation of breast cancer, accompanied with CD36 upregulation. CONCLUSIONS though expression of Sirt1 increased in breast cancer progression, overexpression of Sirt1 could inhibit MCF7 proliferation, which may be associated with CD36 upregulation. In addition, the promotion effect of Dex on MCF7 cells, which may be associated with the Sirt1/CD36 inhibition, could be weakened by Sirt1 activation via RSV.
Collapse
Affiliation(s)
- Yiqun Yao
- Department of Thyroid and Breast Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Tao Liu
- Department of Thyroid and Breast Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Xiaolan Wang
- Department of Thyroid and Breast Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Dianlong Zhang
- Department of Thyroid and Breast Surgery, the Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China.
| |
Collapse
|
16
|
Brecklinghaus T. Highlight report: Import of fatty acids by metastasizing tumor cells. EXCLI JOURNAL 2019; 17:1154-1156. [PMID: 30713475 PMCID: PMC6341421 DOI: 10.17179/excli2018-1870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 11/29/2022]
Affiliation(s)
- Tim Brecklinghaus
- Leibniz Research Centre for Working Environment and Human Factors at the Technical University of Dortmund (IfADo), 44139, Dortmund, Germany
| |
Collapse
|
17
|
Niu B, He K, Li P, Gong J, Zhu X, Ye S, Ou Z, Ren G. SIRT1 upregulation protects against liver injury induced by a HFD through inhibiting CD36 and the NF‑κB pathway in mouse kupffer cells. Mol Med Rep 2018; 18:1609-1615. [PMID: 29845302 PMCID: PMC6072223 DOI: 10.3892/mmr.2018.9088] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 05/17/2018] [Indexed: 11/24/2022] Open
Abstract
Sirtuin 1 (SIRT1) is an NAD(+)-dependent deacetylase, and a critical regulator in various metabolic processes, such as non-alcoholic fatty liver disease (NAFLD). The present study aimed to investigate whether activating SIRT1 could modulate the CD36 and nuclear factor (NF)-κB pathways to protect against liver injury induced by a high-fat diet (HFD) in mice. A mouse NAFLD model was established by administration of a HFD for 8 weeks. During the last 4 weeks, SRT1720, a specific SIRT1 activator, was added daily to the HFD feed. The hepatic morphological structure was observed using hematoxylin and eosin staining, and the ultrastructures in the liver tissue were observed using transmission electron microscopy. Protein expression of SIRT1, CD36 and P65 in liver tissues was detected by immunohistochemistry. Kupffer cells (KCs) from the livers of the mouse models were isolated to determine the mRNA and protein expression of SIRT1, CD36 and P65. SIRT1 activation attenuated the HFD-induced liver injury and significantly reduced the body weight and the levels of alanine transaminase, aspartate aminotransferase, triglyceride, tumor necrosis factor-α and interleukin-6. We observed an increased expression of SIRT1 in the liver tissues from the HFD+SRT1720 group compared with the HFD group. Simultaneously, the expression of CD36 and P65 in the liver tissues was downregulated in the HFD+SRT1720 group. The mRNA and protein expression of SIRT1 was elevated in the HFD+SRT1720 group, whereas the mRNA and protein expression of CD36 and P65 in KCs was significantly decreased in the HFD+SRT1720 group. The present study demonstrated that SIRT1 activation attenuated HFD-induced liver steatosis and inflammation by inhibiting CD36 expression and the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Bailin Niu
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Kun He
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Peizhi Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jianping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xiwen Zhu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Shangmin Ye
- Department of Emergency, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Zhibing Ou
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Guosheng Ren
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
18
|
Chen YP, Kuo WW, Baskaran R, Day CH, Chen RJ, Wen SY, Ho TJ, Padma VV, Kuo CH, Huang CY. Acute hypoxic preconditioning prevents palmitic acid-induced cardiomyocyte apoptosis via switching metabolic GLUT4-glucose pathway back to CD36-fatty acid dependent. J Cell Biochem 2018; 119:3363-3372. [PMID: 29130531 DOI: 10.1002/jcb.26501] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is a risk factor for the development of cardiovascular diseases. Myocardial cell damage leads to an imbalance of energy metabolism, and many studies have indicated that short-term hypoxia during myocardial cell injury has a protective effect. In our previous animal studies, we found that short-term hypoxia in the heart has a protective effect, but long-term hypoxia increases myocardial cell injury. Palmitic acid (PA)-treated H9c2 cardiomyoblasts and neonatal rat ventricle cardiomyocytes were used to simulate hyperlipidemia model, which suppress cluster of differentiation 36 (CD36) and activate glucose transporter type 4 (GLUT4). We exposed the cells to short- and long-term hypoxia and investigated the protective effects of hypoxic preconditioning on PA-induced lipotoxicity in H9c2 cardiomyoblasts and neonatal rat cardiomyocytes. Preconditioning with short-term hypoxia enhanced both CD36 and GLUT4 metabolism pathway protein levels. Expression levels of phospho-PI3K, phospho-Akt, phospho-AMPK, SIRT1, PGC1α, PPARα, CD36, and CPT1β induced by PA was reversed by short-term hypoxia in a time-dependent manner. PA-induced increased GLUT4 membrane protein level was reduced in the cells exposed to short-term hypoxia and si-PKCζ. Short-term hypoxia, resveratrol and si-PKCζ rescue H9c2 cells from apoptosis induced by PA and switch the metabolic pathway from GLUT4 dependent to CD36 dependent. We demonstrate short-term hypoxic preconditioning as a more efficient way as resveratrol in maintaining the energy metabolism of hearts during hyperlipidemia and can be used as a therapeutic strategy.
Collapse
Affiliation(s)
- Yeh-Peng Chen
- PhD Program for Aging, China Medical University, Taichung, Taiwan.,Division of Cardiology, Department of Internal Medicine, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Rathinasamy Baskaran
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | | | - Ray-Jade Chen
- Department of Surgery, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
| | - Tsung-Jung Ho
- Department of Chinese Medicine, China Medical University Beigang Hospital, Taichung, Taiwan.,Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
| | | | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
19
|
Sun T, Zhang Y, Zhong S, Gao F, Chen Y, Wang B, Cai W, Zhang Z, Li W, Lu S, Zheng F, Shi G. N-n-Butyl Haloperidol Iodide, a Derivative of the Anti-psychotic Haloperidol, Antagonizes Hypoxia/Reoxygenation Injury by Inhibiting an Egr-1/ROS Positive Feedback Loop in H9c2 Cells. Front Pharmacol 2018; 9:19. [PMID: 29422863 PMCID: PMC5789774 DOI: 10.3389/fphar.2018.00019] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/08/2018] [Indexed: 02/05/2023] Open
Abstract
Early growth response-1 (Egr-1), a transcription factor which often underlies the molecular basis of myocardial ischemia/reperfusion (I/R) injury, and oxidative stress, is key to myocardial I/R injury. Silent information regulator of transcription 1(SIRT1) not only interacts with and is inhibited by Egr-1, but also downregulates reactive oxygen species (ROS) via the Forkhead box O1(FOXO1)/manganese superoxide dismutase (Mn-SOD) signaling pathway. N-n-butyl haloperidol iodide (F2), a new patented compound, protects the myocardium against myocardial I/R injury in various animal I/R models in vivo and various heart-derived cell hypoxia/reoxygenation (H/R) models in vitro. In addition, F2 can regulate the abnormal ROS/Egr-1 signaling pathway in cardiac microvascular endothelial cells (CMECs) and H9c2 cells after H/R. We studied whether there is an inverse Egr-1/ROS signaling pathway in H9c2 cells and whether the SIRT1/FOXO1/Mn-SOD signaling pathway mediates this. We verified a ROS/Egr-1 signaling loop in H9c2 cells during H/R and that F2 protects against myocardial H/R injury by affecting SIRT1-related signaling pathways. Knockdown of Egr-1, by siRNA interference, reduced ROS generation, and alleviated oxidative stress injury induced by H/R, as shown by upregulated mitochondrial membrane potential, increased glutathione peroxidase (GSH-px) and total SOD anti-oxidative enzyme activity, and downregulated MDA. Decreases in FOXO1 protein expression and Mn-SOD activity occurred after H/R, but could be blocked by Egr-1 siRNA. F2 treatment attenuated H/R-induced Egr-1 expression, ROS generation and other forms of oxidative stress injury such as MDA, and prevented H/R-induced decreases in FOXO1 and Mn-SOD activity. Nuclear co-localization between Egr-1 and SIRT1 was increased by H/R and decreased by either Egr-1 siRNA or F2. Therefore, our results suggest that Egr-1 inhibits the SIRT1/FOXO1/Mn-SOD antioxidant signaling pathway to increase ROS and perpetuate I/R injury. F2 inhibits induction of Egr-1 by H/R, thereby activating SIRT1/FOXO1/Mn-SOD antioxidant signaling and decreasing H/R-induced ROS, demonstrating an important mechanism by which F2 protects against myocardial H/R injury.
Collapse
Affiliation(s)
- Ting Sun
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Yanmei Zhang
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Shuping Zhong
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, CA, United States
| | - Fenfei Gao
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Yicun Chen
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Bin Wang
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Wenfeng Cai
- Department of Pharmacology, Shantou University Medical College, Shantou, China
| | - Zhaojing Zhang
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Weiqiu Li
- Analytical Cytology Laboratory, Shantou University Medical College, Shantou, China
| | - Shishi Lu
- Department of Pharmacy, The First Affiliated Hospital, Shantou University Medical College, Shantou, China
| | - Fuchun Zheng
- Clinical Pharmacology Laboratory, The First Affiliated Hospital, Shantou University Medical College, Shantou, China
- *Correspondence: Ganggang Shi, Fuchun Zheng,
| | - Ganggang Shi
- Department of Pharmacology, Shantou University Medical College, Shantou, China
- *Correspondence: Ganggang Shi, Fuchun Zheng,
| |
Collapse
|
20
|
Padmanabhan S, Joe B. Towards Precision Medicine for Hypertension: A Review of Genomic, Epigenomic, and Microbiomic Effects on Blood Pressure in Experimental Rat Models and Humans. Physiol Rev 2017; 97:1469-1528. [PMID: 28931564 PMCID: PMC6347103 DOI: 10.1152/physrev.00035.2016] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 04/28/2017] [Accepted: 04/29/2017] [Indexed: 12/11/2022] Open
Abstract
Compelling evidence for the inherited nature of essential hypertension has led to extensive research in rats and humans. Rats have served as the primary model for research on the genetics of hypertension resulting in identification of genomic regions that are causally associated with hypertension. In more recent times, genome-wide studies in humans have also begun to improve our understanding of the inheritance of polygenic forms of hypertension. Based on the chronological progression of research into the genetics of hypertension as the "structural backbone," this review catalogs and discusses the rat and human genetic elements mapped and implicated in blood pressure regulation. Furthermore, the knowledge gained from these genetic studies that provide evidence to suggest that much of the genetic influence on hypertension residing within noncoding elements of our DNA and operating through pervasive epistasis or gene-gene interactions is highlighted. Lastly, perspectives on current thinking that the more complex "triad" of the genome, epigenome, and the microbiome operating to influence the inheritance of hypertension, is documented. Overall, the collective knowledge gained from rats and humans is disappointing in the sense that major hypertension-causing genes as targets for clinical management of essential hypertension may not be a clinical reality. On the other hand, the realization that the polygenic nature of hypertension prevents any single locus from being a relevant clinical target for all humans directs future studies on the genetics of hypertension towards an individualized genomic approach.
Collapse
Affiliation(s)
- Sandosh Padmanabhan
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; and Center for Hypertension and Personalized Medicine; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| | - Bina Joe
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; and Center for Hypertension and Personalized Medicine; Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, Ohio
| |
Collapse
|
21
|
Wen SY, Velmurugan BK, Day CH, Shen CY, Chun LC, Tsai YC, Lin YM, Chen RJ, Kuo CH, Huang CY. High density lipoprotein (HDL) reverses palmitic acid induced energy metabolism imbalance by switching CD36 and GLUT4 signaling pathways in cardiomyocyte. J Cell Physiol 2017; 232:3020-3029. [PMID: 28500736 DOI: 10.1002/jcp.26007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 05/12/2017] [Indexed: 01/16/2023]
Abstract
In our previous study palmitic acid (PA) induced lipotoxicity and switches energy metabolism from CD36 to GLUT4 in H9c2 cells. Low level of high density lipoprotein (HDL) is an independent risk factor for cardiac hypertrophy. Therefore, we in the present study investigated whether HDL can reverse PA induced lipotoxicity in H9c2 cardiomyoblast cells. In this study, we treated H9c2 cells with PA to create a hyperlipidemia model in vitro and analyzed for CD36 and GLUT4 metabolic pathway proteins. CD36 metabolic pathway proteins (phospho-AMPK, SIRT1, PGC1α, PPARα, CPT1β, and CD36) were decreased by high PA (150 and 200 μg/μl) concentration. Interestingly, expression of GLUT4 metabolic pathway proteins (p-PI3K and pAKT) were increased at low concentration (50 μg/μl) and decreased at high PA concentration. Whereas, phospho-PKCζ, GLUT4 and PDH proteins expression was increased in a dose dependent manner. PA treated H9c2 cells were treated with HDL and analyzed for cell viability. Results showed that HDL treatment induced cell proliferation efficiency in PA treated cells. In addition, HDL reversed the metabolic effects of PA: CD36 translocation was increased and reduced GLUT4 translocation, but HDL treatment significantly increased CD36 metabolic pathway proteins and reduced GLUT4 pathway proteins. Rat neonatal cardiomyocytes showed similar results. In conclusion, HDL reversed palmatic acid-induced lipotoxicity and energy metabolism imbalance in H9c2 cardiomyoblast cells and in neonatal rat cardiomyocyte cells.
Collapse
Affiliation(s)
- Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan.,Center for General Education, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| | | | | | - Chia-Yao Shen
- Department of Nursing, MeiHo University, Pingtung, Taiwan
| | - Li-Chin Chun
- Department of Hospital and Health Care Administration, Chia Nan University of Pharmacy and Science, Tainan County, Taiwan
| | - Yi-Chieh Tsai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan.,Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.,Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan.,Department of Biological Science and Technology, Asia University, Taichung, Taiwan
| |
Collapse
|