1
|
Wang T, Wang YY, Shi MY, Liu L. Mechanisms of action of natural products on type 2 diabetes. World J Diabetes 2023; 14:1603-1620. [DOI: 10.4239/wjd.v14.i11.1603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/14/2023] [Accepted: 10/23/2023] [Indexed: 11/14/2023] Open
Abstract
Over the past several decades, type 2 diabetes mellitus (T2DM) has been considered a global public health concern. Currently, various therapeutic modalities are available for T2DM management, including dietary modifications, moderate exercise, and use of hypoglycemic agents and lipid-lowering medications. Although the curative effect of most drugs on T2DM is significant, they also exert some adverse side effects. Biologically active substances found in natural medicines are important for T2DM treatment. Several recent studies have reported that active ingredients derived from traditional medicines or foods exert a therapeutic effect on T2DM. This review compiled important articles regarding the therapeutic effects of natural products and their active ingredients on islet β cell function, adipose tissue inflammation, and insulin resistance. Additionally, this review provided an in-depth understanding of the multiple regulatory effects on different targets and signaling pathways of natural medicines in the treatment of T2DM as well as a theoretical basis for clinical effective application.
Collapse
Affiliation(s)
- Tao Wang
- Clinical Molecular Immunology Center, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Yang-Yang Wang
- Clinical Molecular Immunology Center, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Meng-Yue Shi
- Clinical Molecular Immunology Center, Yangtze University, Jingzhou 434023, Hubei Province, China
| | - Lian Liu
- Department of Pharmacology, Yangtze University, Jingzhou 434023, Hubei Province, China
| |
Collapse
|
2
|
Gao S, Feng Q. The Beneficial Effects of Geniposide on Glucose and Lipid Metabolism: A Review. Drug Des Devel Ther 2022; 16:3365-3383. [PMID: 36213380 PMCID: PMC9532263 DOI: 10.2147/dddt.s378976] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Geniposide is a naturally sourced active ingredient that has diverse pharmacological effects and great potential in improving or treating different kinds of diseases. In recent years, more and more studies have confirmed that geniposide can improve glucose and lipid metabolism disorder, which is an increasingly prevalent health problem causing various metabolic diseases globally. Our review aims to summarize basic information on the pharmacological effects of geniposide on glucolipid metabolism. Geniposide increases glucose utilization and insulin production, protects pancreatic islet β cells, inhibits insulin resistance and hepatic glucose production, and suppresses gluconeogenesis. While in the aspect of lipid metabolism, geniposide can promote lipolysis, inhibit lipogenesis, and regulate lipid transport. Geniposide ameliorates lipid and glucose metabolic disorders, improving the entire glycolipid metabolism network in a three-dimensional manner at the level of molecular mechanism. Growing evidence revealed that geniposide may serve as an effective drug to combat metabolic diseases for the time to come.
Collapse
Affiliation(s)
- Siting Gao
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Hepatopathy Building, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Qin Feng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Hepatopathy Building, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| |
Collapse
|
3
|
Hong H, Xu J, He H, Wang X, Yang L, Deng P, Yang L, Tan M, Zhang J, Xu Y, Tong T, Lin X, Pi H, Lu Y, Zhou Z. Cadmium perturbed metabolomic signature in pancreatic beta cells correlates with disturbed metabolite profile in human urine. ENVIRONMENT INTERNATIONAL 2022; 161:107139. [PMID: 35172228 DOI: 10.1016/j.envint.2022.107139] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 02/04/2022] [Accepted: 02/05/2022] [Indexed: 06/14/2023]
Abstract
Cd exposure has been demonstrated to induce a variety of metabolic disorders accompanied with imbalance of glucose and lipid homeostasis. The metabolic toxicity of Cd exposure at metabolome-wide level remains elusive. In our study, we demonstrated that Cd exposure via drinking water increased blood glucose levels, decreased serum insulin levels, led to glucose intolerance and suppressed insulin expression in the pancreas of C57/6J mice. Cd exposure significantly inhibited cell viability and suppressed insulin secretion in MIN6 cells in vitro. Since pancreatic β-cells are the only source of insulin production in the body and play a pivotal role in modulating glucose and lipid metabolisms, we further delineated the metabolomic signatures of Cd exposure in insulin-secreting MIN6 cells by using non-target metabolomics. PCA and OPLS-DA analysis clearly suggested that Cd exposure led to a marked metabolic alteration in MIN6 cells. 76 perturbed metabolites were identified after Cd exposure. Classification of metabolites suggested that Cd perturbed metabolites belong to nucleosides, nucleotides and analogues, organic acids and derivatives, and lipids and lipid-like molecules. 28 perturbed metabolites existed in mitochondrion, suggesting mitochondrion as the major target organelle in metabolic toxicity of Cd exposure. KEGG pathway analysis revealed that 20 metabolic pathways were disturbed by Cd exposure. Mitochondrial TCA cycle and glycerophospholipid metabolism were remarkably disturbed. The mRNA expressions of genes in mitochondrial TCA cycle and fatty acid oxidation in pancreas and MIN6 cells were significantly dysregulated by Cd exposure. Disturbances in mitochondrial TCA cycle and glycerophospholipid metabolism result in producing perturbed metabolites in pancreatic β-cells. Moreover, 14 perturbed metabolites identified in MIN6 cells co-existed in the urine of Cd exposed workers. 11 biomarkers of diabetes mellitus were also found to be significantly altered in the urine of Cd exposed workers. In conclusion, findings of this study greatly extend our understanding of metabolic toxicity of Cd exposure in pancreatic β-cells at metabolome-wide level and offer some new clues for linking Cd exposure to development of diabetes mellitus. Results of this study also support the notion that Cd induced metabolic toxicity could be monitored by examining perturbed urinary metabolites in humans and highlight the significance of reducing Cd exposure via drinking water at population level.
Collapse
Affiliation(s)
- Huihui Hong
- Department of Emergency Medicine, The First Affiliated Hospital and Department of Environmental Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jia Xu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haotian He
- Department of Environmental Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xue Wang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Lingling Yang
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Ping Deng
- Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Lu Yang
- Hunan Province Prevention and Treatment Hospital for Occupational Diseases, Hunan, China
| | - Miduo Tan
- Department of Galactophore, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Jingjing Zhang
- Department of Environmental Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yudong Xu
- Department of Environmental Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Tong Tong
- Department of Environmental Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiqin Lin
- Department of Environmental Medicine, School of Medicine, Zhejiang University, Hangzhou, China
| | - Huifeng Pi
- Department of Occupational Health, Third Military Medical University, Chongqing, China.
| | - Yuanqiang Lu
- Department of Emergency Medicine, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Key Laboratory of Diagnosis and Treatment of Aging and Physic-chemical Injury Diseases of Zhejiang Province, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhou Zhou
- Department of Emergency Medicine, The First Affiliated Hospital and Department of Environmental Medicine, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Drzazga A, Kamińska D, Gliszczyńska A, Gendaszewska-Darmach E. Isoprenoid Derivatives of Lysophosphatidylcholines Enhance Insulin and GLP-1 Secretion through Lipid-Binding GPCRs. Int J Mol Sci 2021; 22:5748. [PMID: 34072220 PMCID: PMC8197866 DOI: 10.3390/ijms22115748] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/13/2022] Open
Abstract
Insulin plays a significant role in carbohydrate homeostasis as the blood glucose lowering hormone. Glucose-induced insulin secretion (GSIS) is augmented by glucagon-like peptide (GLP-1), a gastrointestinal peptide released in response to ingesting nutriments. The secretion of insulin and GLP-1 is mediated by the binding of nutrients to G protein-coupled receptors (GPCRs) expressed by pancreatic β-cells and enteroendocrine cells, respectively. Therefore, insulin secretagogues and incretin mimetics currently serve as antidiabetic treatments. This study demonstrates the potency of synthetic isoprenoid derivatives of lysophosphatidylcholines (LPCs) to stimulate GSIS and GLP-1 release. Murine insulinoma cell line (MIN6) and enteroendocrinal L cells (GLUTag) were incubated with LPCs bearing geranic acid (1-GA-LPC), citronellic acid (1-CA-LPC), 3,7-dimethyl-3-vinyloct-6-enoic acid (GERA-LPC), and (E)-3,7,11-trimethyl- 3-vinyldodeca-6,10-dienoic acid (1-FARA-LPC). Respective free terpene acids were also tested for comparison. Besides their insulin- and GLP-1-secreting capabilities, we also investigated the cytotoxicity of tested compounds, the ability to intracellular calcium ion mobilization, and targeted GPCRs involved in maintaining lipid and carbohydrate homeostasis. We observed the high cytotoxicity of 1-GERA-LPC and 1-FARA-LPC in contrast 1-CA-LPC and 1-GA-LPC. Moreover, 1-CA-LPC and 1-GA-LPC demonstrated the stimulatory effect on GSIS and 1-CA-LPC augmented GLP-1 secretion. Insulin and GLP-1 release appeared to be GPR40-, GPR55-, GPR119- and GPR120-dependent.
Collapse
Affiliation(s)
- Anna Drzazga
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (A.D.); (D.K.)
| | - Daria Kamińska
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (A.D.); (D.K.)
| | - Anna Gliszczyńska
- Department of Chemistry, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375 Wrocław, Poland
| | - Edyta Gendaszewska-Darmach
- Institute of Molecular and Industrial Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Stefanowskiego 4/10, 90-924 Lodz, Poland; (A.D.); (D.K.)
| |
Collapse
|
5
|
Zhao W, Pu M, Shen S, Yin F. Geniposide improves insulin resistance through AMPK-mediated Txnip protein degradation in 3T3-L1 adipocytes. Acta Biochim Biophys Sin (Shanghai) 2021; 53:160-169. [PMID: 33349852 DOI: 10.1093/abbs/gmaa157] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Indexed: 12/29/2022] Open
Abstract
Thioredoxin-interacting protein (Txnip) has emerged as a key regulator of insulin resistance. In this study, we investigated the roles of geniposide and Txnip in insulin resistance in differentiated 3T3-L1 adipocytes. Our results revealed that geniposide markedly enhanced glucose uptake, increased the protein levels of insulin receptor substrate (IRS)-1 and GLUT-1, and prevented the phosphorylation of IRS-1 and Akt Thr308 induced by insulin resistance in 3T3-L1 adipocytes. We also observed that geniposide accelerated protein degradation of Txnip through proteasome pathway, and knockdown of Txnip with small interfering RNA attenuated the effect of geniposide on insulin signaling molecules, implying that Txnip played a pivotal role in the regulation of insulin signaling molecules by geniposide in 3T3-L1 adipocytes. Furthermore, geniposide induced the phosphorylation of adenosine monophosphate-activated protein kinase (AMPK) in the presence of high glucose in differentiated 3T3-L1 adipocytes, while compound C, an inhibitor of AMPK, prevented the effect of geniposide on Txnip degradation and the regulation of glucose uptake and insulin signaling molecules including p-IRS-1, IRS-1, and GLUT-1 in differentiated 3T3-L1 adipocytes. Taken together, all these findings suggest that geniposide improves the insulin signaling defect possibly by AMPK-mediated Txnip degradation in 3T3-L1 adipocytes.
Collapse
Affiliation(s)
- Wanjun Zhao
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology of Technology, Chongqing University of Technology, Chongqing 400054, China
| | - Mengru Pu
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology of Technology, Chongqing University of Technology, Chongqing 400054, China
| | - Shenli Shen
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology of Technology, Chongqing University of Technology, Chongqing 400054, China
| | - Fei Yin
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology of Technology, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
6
|
dos Santos FB, Quines CB, Pilissão LEB, Dal Forno AHDC, Rodrigues CF, Denardin CC, Farias FM, Ávila DS. Aqueous Bark Extract of Ceiba speciosa (A. St.-Hill) Ravenna Protects against Glucose Toxicity in Caenorhabditis elegans. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1321354. [PMID: 33101582 PMCID: PMC7568133 DOI: 10.1155/2020/1321354] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/01/2020] [Accepted: 07/25/2020] [Indexed: 12/25/2022]
Abstract
Plants are widely used in folk medicine because of their pharmacological properties. Ceiba speciosa, popularly known as paineira-rosa or tree-of-wool, is a species found in the Northwest of Rio Grande do Sul, being native of the upper Uruguay River, Brazil. The tea obtained from the stem bark is employed in folk medicine to reduce cholesterol, triacylglycerides, and glucose levels. However, there are no studies in the literature proving its efficacy or the safety of its use. For this study, we used Caenorhabditis elegans as an animal model considering its advantages for risk assessment and pharmacological screenings. For the toxicological tests, C. elegans N2 (wild type) was treated with the aqueous extract of the stem bark of C. speciosa (ECE) at the first larval stage (L1) at concentrations of 5, 25, 50, and 250 μg/mL. To evaluate biological activities, we challenged the extract for oxidative stress resistance in the presence of paraquat (0.5 mM), H2O2 (1 mM), and against glucose-induced toxicity. Our results demonstrated that ECE did not alter survival rate, pharyngeal pumping, and reproduction of the nematodes. The extract was not able to protect the nematodes against the toxicity induced by prooxidants. Notably, ECE protected against glucotoxicity by increasing worms' life span and by reducing glucose levels. On the other hand, ECE treatment did not reduce lipid accumulation induced by exogenous glucose feeding, as observed in worms which lipid droplets were tagged with GFP. Based on our results, we believe that the extract is indeed promising for further studies focusing on carbohydrates metabolism; however, it needs to be carefully evaluated since the extract does not seem to modulate lipid accumulation.
Collapse
Affiliation(s)
- Fabrine Bianchin dos Santos
- Laboratório de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| | - Caroline Brandão Quines
- Laboratório de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| | - Luiz Eduardo Ben Pilissão
- Laboratório de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| | - Ana Helena de Castro Dal Forno
- Laboratório de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| | - Cristiane Freitas Rodrigues
- Laboratório de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| | - Cristiane Casagrande Denardin
- Laboratório de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| | - Fabiane Moreira Farias
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| | - Daiana Silva Ávila
- Laboratório de Pesquisa em Bioquímica e Toxicologia em Caenorhabditis elegans, Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa, Uruguaiana, Brazil BR 472-Km 592-Caixa Postal 118, CEP 97500-970
| |
Collapse
|
7
|
Cui LJ, Bai T, Zhi LP, Liu ZH, Liu T, Xue H, Yang HH, Yang XH, Zhang M, Niu YR, Liu YF, Zhang Y. Analysis of long noncoding RNA-associated competing endogenous RNA network in glucagon-like peptide-1 receptor agonist-mediated protection in β cells. World J Diabetes 2020; 11:374-390. [PMID: 32994866 PMCID: PMC7503504 DOI: 10.4239/wjd.v11.i9.374] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 04/24/2020] [Accepted: 08/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) and mRNAs are widely involved in various physiological and pathological processes. The use of glucagon-like peptide-1 receptor agonists (GLP-1RAs) is a novel therapeutic strategy that could promote insulin secretion and decrease the rate of β-cell apoptosis in type 2 diabetes mellitus (T2DM) patients. However, the specific lncRNAs and mRNAs and their functions in these processes have not been fully identified and elucidated.
AIM To identify the lncRNAs and mRNAs that are involved in the protective effect of GLP-1RA in β cells, and their roles.
METHODS Rat gene microarray was used to screen differentially expressed (DE) lncRNAs and mRNAs in β cells treated with geniposide, a GLP-1RA. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed to assess the underlying functions of DE mRNAs. Hub mRNAs were filtered using the STRING database and the Cytoscape plugin, CytoHubba. In order to reveal the regulatory relationship between lncRNAs and hub mRNAs, their co-expression network was constructed based on the Pearson coefficient of DE lncRNAs and mRNAs, and competing endogenous RNA (ceRNA) mechanism was explored through miRanda and TargetScan databases.
RESULTS We identified 308 DE lncRNAs and 128 DE mRNAs with a fold change filter of ≥ 1.5 and P value < 0.05. GO and KEGG pathway enrichment analyses indicated that the most enriched terms were G-protein coupled receptor signaling pathway, inflammatory response, calcium signaling pathway, positive regulation of cell proliferation, and ERK1 and ERK2 cascade. Pomc, Htr2a, and Agtr1a were screened as hub mRNAs using the STRING database and the Cytoscape plugin, CytoHubba. This result was further verified using SwissTargetPrediction tool. Through the co-expression network and competing endogenous (ceRNA) mechanism, we identified seven lncRNAs (NONRATT027738, NONRATT027888, NONRATT030038, etc.) co-expressed with the three hub mRNAs (Pomc, Htr2a, and Agtr1a) based on the Pearson coefficient of the expression levels. These lncRNAs regulated hub mRNA functions by competing with six miRNAs (rno-miR-5132-3p, rno-miR-344g, rno-miR-3075, etc.) via the ceRNA mechanism. Further analysis indicated that lncRNA NONRATT027738 interacts with all the three hub mRNAs, suggesting that it is at a core position within the ceRNA network.
CONCLUSION We have identified key lncRNAs and mRNAs, and highlighted here how they interact through the ceRNA mechanism to mediate the protective effect of GLP-1RA in β cells.
Collapse
Affiliation(s)
- Li-Juan Cui
- Department of Pharmacology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Tao Bai
- Department of Endocrinology, The First Clinical Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Lin-Ping Zhi
- Department of Pharmacology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Zhi-Hong Liu
- Department of Respiratory Medicine, The First Clinical Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Tao Liu
- Department of General Surgery, Shanxi Bethune Hospital, Taiyuan 030006, Shanxi Province, China
| | - Huan Xue
- Department of Pharmacology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Huan-Huan Yang
- Department of Pharmacology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Xiao-Hua Yang
- Department of Pharmacology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Min Zhang
- College of Pharmacy, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Ya-Ru Niu
- Second Clinical Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yun-Feng Liu
- Department of Endocrinology, The First Clinical Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yi Zhang
- Department of Pharmacology, Basic Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| |
Collapse
|
8
|
The Effects of Natural Iridoids and Anthocyanins on Selected Parameters of Liver and Cardiovascular System Functions. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:2735790. [PMID: 32318236 PMCID: PMC7150688 DOI: 10.1155/2020/2735790] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/10/2020] [Accepted: 03/06/2020] [Indexed: 12/29/2022]
Abstract
The old adage says, "you are what you eat." And although it is a banality repeated by many with a grain of salt, it also has quite a bit of truth in it, as the products we eat have a considerable impact on our health. Unfortunately, humanity is eating worse from one year to another, both in terms of product quality and eating habits. At the same time, it is brought up frequently that plant products should form the basis of our diet. This issue was also reflected in the new version of the food pyramid. Iridoids and anthocyanins are groups of plant compounds with proven beneficial effects on health. Both groups affect the cardiovascular system and the liver functions. Although many mechanisms of action and the therapeutic effects of these compounds have already been learned, intensive animal and clinical research is still underway to explore their new curative mechanisms and effects or to broaden our knowledge of those previously described. In this article, we review the effects of natural iridoids and anthocyanins on selected parameters of liver and cardiovascular system functions.
Collapse
|
9
|
Liu M, Liu C, Shen S, Liu J, Yin F. Geniposide inhibits glucolipotoxicity and cooperates with Txnip knockdown to potentiate cell adaption to endoplasmic reticulum stress in pancreatic beta cells. Cell Biol Int 2020; 44:1535-1543. [PMID: 32215982 DOI: 10.1002/cbin.11350] [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: 10/02/2019] [Revised: 03/09/2020] [Accepted: 03/19/2020] [Indexed: 11/09/2022]
Abstract
Thioredoxin-interacting protein (Txnip), a negative regulator of thioredoxin, has become an attractive therapeutic target to alleviate metabolic diseases. Our previous data demonstrated that geniposide improved glucose-stimulated insulin secretion by accelerating Txnip degradation and prevented the early-stage apoptosis of pancreatic β cells induced by palmitate, but the underlying mechanisms are still unclear. The objective of this study is to identify the role of Txnip in geniposide preventing the apoptosis of pancreatic β cells induced by high glucose and palmitate (HG/PA). The results revealed that geniposide attenuated HG/PA-induced cell apoptosis and the expression of Bax and caspase-3, while increasing mitochondrial membrane potential and the anti-apoptotic protein levels of heme-oxygenase-1 (HO-1) and Bcl-2 in INS-1 rat pancreatic β cells. Knockdown of the Txnip gene raised the levels of anti-apoptotic proteins HO-1 and Bcl-2 and geniposide potentiated the effect of Txnip when the INS-1 cells were challenged by HG/PA. Furthermore, geniposide enhanced the adoptive unfolded protein response by increasing the phosphorylation of PERK/eIF2α and IRE1α in HG/PA-treated INS-1 cells. The results together suggest that geniposide might be useful to antagonize glucolipotoxicity and Txnip might be a pleiotropic cellular factor in pancreatic β cells.
Collapse
Affiliation(s)
- Min Liu
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Chunyan Liu
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Shenli Shen
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Jianhui Liu
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| | - Fei Yin
- Chongqing Key Lab of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing, 400054, China
| |
Collapse
|
10
|
Zhang X, Gao T, Wang Y. Geniposide alleviates lipopolysaccharide (LPS)-induced inflammation by downregulation of miR-27a in rat pancreatic acinar cell AR42J. Biol Chem 2019; 400:1059-1068. [PMID: 30897061 DOI: 10.1515/hsz-2018-0422] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 03/15/2019] [Indexed: 01/17/2023]
Abstract
Pancreatitis is a disease caused by inflammation of pancreatic acinar cells. Geniposide (GEN) possesses anti-inflammation activities. Hence, we investigated the effects of GEN on lipopolysaccharide (LPS)-stimulated AR42J cells. AR42J cells were stimulated by LPS and then treated with GEN and/or transfected with miR-27a mimic or negative control. Cell viability and cell apoptosis were detected using the Cell Counting Kit-8 and flow cytometry, respectively. All related proteins were measured by Western blot. The expression of miR-27a was detected by quantitative real time-polymerase chain reaction (qRT-PCR). Moreover, the expression of inflammatory cytokines interleukin-6 (IL-6) and monocyte chemoattractant protein (MCP)-1 was analyzed by qRT-PCR and Western blot. LPS significantly decreased cell viability, and enhanced cell apoptosis and IL-6, MCP-1 expression. Then GEN administration alleviated inflammatory injury by increasing cell viability, while reducing apoptosis, and IL-6 and MCP-1 expression. GEN downregulated miR-27a expression which was induced by LPS. Transfection with miR-27a mimic partially eliminated the protective effects of GEN. The phosphorylation of JNK and c-Jun was downregulated by GEN while upregulated by miR-27a overexpression. GEN alleviates LPS-induced AR42J cell injury as evidenced by promoting cell growth, and upregulation of IL-6 and MCP-1. This process might be modulated by down-regulating miR-27a and inactivation of JNK pathway.
Collapse
Affiliation(s)
- Xiaofen Zhang
- Department of Critical Care Medicine, Jining No. 1 People's Hospital, No. 6 Jiankang Road, Jining 272000, China
- Affiliated Jining No. 1 People's Hospital of Jining Medical University, Jining Medical University, Jining 272000, China
| | - Taishan Gao
- Department of Critical Care Medicine, Jining No. 1 People's Hospital, No. 6 Jiankang Road, Jining 272000, China
| | - Yanhua Wang
- Department of Critical Care Medicine, Jining No. 1 People's Hospital, No. 6 Jiankang Road, Jining 272000, China
| |
Collapse
|
11
|
Hao Y, Shen S, Yin F, Zhang Y, Liu J. Unfolded protein response is involved in geniposide‐regulating glucose‐stimulated insulin secretion in INS‐1 cells. Cell Biochem Funct 2019; 37:368-376. [DOI: 10.1002/cbf.3414] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/06/2019] [Indexed: 02/04/2023]
Affiliation(s)
- Yanan Hao
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Shenli Shen
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Fei Yin
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Yonglan Zhang
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| | - Jianhui Liu
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular PharmacologyChongqing University of Technology Chongqing China
| |
Collapse
|
12
|
Xie B, Wu J, Li Y, Wu X, Zeng Z, Zhou C, Xu D, Wu L. Geniposide Alleviates Glucocorticoid-Induced Inhibition of Osteogenic Differentiation in MC3T3-E1 Cells by ERK Pathway. Front Pharmacol 2019; 10:411. [PMID: 31057410 PMCID: PMC6482204 DOI: 10.3389/fphar.2019.00411] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 04/01/2019] [Indexed: 12/12/2022] Open
Abstract
Glucocorticoid (GC) therapy is the leading cause of secondary osteoporosis and the therapeutic and preventative drugs for GC-induced osteoporosis are limited. In this study, we investigated the protective effects of geniposide on dexamethasone (DEX)-induced osteogenic inhibition in MC3T3-E1 cells. The results showed that there was no obvious toxicity on MC3T3-E1 cells when geniposide was used at the doses ranging from 1 to 75 μM. In DEX-treated MC3T3-E1 cells, geniposide promoted the alkaline phosphatase (ALP) activity and the mineralization. In addition, geniposide also significantly increased the mRNA and protein expression of osteopontin (OPN), Runt-related transcription factor 2 (Runx2), and Osterix (Osx) in DEX-treated MC3T3-E1 cells. Furthermore, geniposide activated ERK pathway in DEX-treated MC3T3-E1 cells. The ERK activation inhibitor U0126 and glucagon-like peptide-1 (GLP-1) receptor antagonist exendin 9-39 abolished the geniposide-induced activation of ERK and inhibited the protective effect of geniposide. Taken together, our study revealed that geniposide alleviated GC-induced osteogenic suppression in MC3T3-E1 cells. The effect of geniposide was at least partially associated with activating ERK signaling pathway via GLP-1 receptor. Geniposide might be a potential therapeutic agent for GC-induced osteoporosis.
Collapse
Affiliation(s)
- Baocheng Xie
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Jiahuan Wu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Yongmei Li
- Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Xuejun Wu
- Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Zhanwei Zeng
- Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Chenhui Zhou
- School of Nursing, Guangdong Medical University, Dongguan, China
| | - Daohua Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.,Department of Pharmacology, Institute of Traditional Chinese Medicine and New Pharmacy Development, Guangdong Medical University, Dongguan, China
| | - Longhuo Wu
- College of Pharmacy, Gannan Medical University, Ganzhou, China
| |
Collapse
|
13
|
Diverse Pharmacological Activities and Potential Medicinal Benefits of Geniposide. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:4925682. [PMID: 31118959 PMCID: PMC6500620 DOI: 10.1155/2019/4925682] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 03/19/2019] [Indexed: 12/25/2022]
Abstract
Geniposide is a well-known iridoid glycoside compound and is an essential component of a wide variety of traditional phytomedicines, for example, Gardenia jasminoides Elli (Zhizi in Chinese), Eucommia ulmoides Oliv. (Duzhong in Chinese), Rehmannia glutinosa Libosch. (Dihuang in Chinese), and Achyranthes bidentata Bl. (Niuxi in Chinese). It is also the main bioactive component of Gardeniae Fructus, the dried ripe fruit of Gardenia jasminoides Ellis. Increasing pharmacological evidence supports multiple medicinal properties of geniposide including neuroprotective, antidiabetic, hepatoprotective, anti-inflammatory, analgesic, antidepressant-like, cardioprotective, antioxidant, immune-regulatory, antithrombotic, and antitumoral effects. It has been proposed that geniposide may be a drug or lead compound for the prophylaxis and treatment of several diseases, such as Alzheimer's disease, Parkinson's disease, diabetes and diabetic complications, ischemia and reperfusion injury, and hepatic disorders. The aim of the present review is to give a comprehensive summary and analysis of the pharmacological properties of geniposide, supporting its use as a medicinal agent.
Collapse
|
14
|
Ren L, Sun D, Zhou X, Yang Y, Huang X, Li Y, Wang C, Li Y. Chronic treatment with the modified Longdan Xiegan Tang attenuates olanzapine-induced fatty liver in rats by regulating hepatic de novo lipogenesis and fatty acid beta-oxidation-associated gene expression mediated by SREBP-1c, PPAR-alpha and AMPK-alpha. JOURNAL OF ETHNOPHARMACOLOGY 2019; 232:176-187. [PMID: 30590197 DOI: 10.1016/j.jep.2018.12.034] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The modified Longdan Xiegan Tang (mLXT) has been used clinically for various neuropsychiatric disorders and liver diseases. The use of antipsychotics is associated with nonalcoholic fatty liver disease. AIM OF THE STUDY To investigate the effect and underlying mechanisms of mLXT on antipsychotic-induced fatty liver. MATERIALS AND METHODS The representative active components in the formula were identified and quantified by a HPLC method. Fatty liver in male rats was induced by olanzapine (5 mg/kg) (p.o., × 8 weeks), and the rats were co-treated with mLXT extract (50 and 500 mg/kg). Blood and liver variables were determined enzymatically or histologically. Gene/protein expression was analyzed by real-time PCR and Western blot. RESULTS Treatment of rats with mLXT decreased olanzapine-induced increases in hepatic triglyceride content, cell vacuolar degeneration and Oil Red O-stained area, accompanied by suppression of olanzapine-stimulated hepatic mRNA and/or protein overexpression of sterol regulatory element-binding protein (SREBP)-1c, and its downstream lipogenic enzymes for de novo lipogenesis. Besides, mLXT also activated hepatic expression of peroxisome proliferator-activated receptor-alpha and its target genes associated with fatty acid beta-oxidation, phosphorylated Thr172 in AMP-activated protein kinase (AMPK)-alpha (the upstream enzyme of SREBP-1c and PPAR-alpha), and its ratio to total AMPK-alpha. CONCLUSIONS The present results suggest that chronic treatment with mLXT ameliorates olanzapine-induced fatty liver by regulating hepatic de novo lipogenesis- and fatty acid beta-oxidation-associated gene expression mediated by SREBP-1c and PPAR-alpha, respectively, through activation of AMPK-alpha. Our findings provide the evidence that supports clinical use of the formula for antipsychotic medication-induced fatty liver.
Collapse
Affiliation(s)
- Liying Ren
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Dongmei Sun
- Analysis Department of Chinese Medicine, Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou 510095, China.
| | - Xia Zhou
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yifan Yang
- Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, NSW 2000, Australia.
| | - Xiaoqian Huang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yangxue Li
- Analysis Department of Chinese Medicine, Guangdong Province Engineering Technology Research Institute of Traditional Chinese Medicine, Guangzhou 510095, China.
| | - Chunxia Wang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Yuhao Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; Endocrinology and Metabolism Group, Sydney Institute of Health Sciences/Sydney Institute of Traditional Chinese Medicine, Sydney, NSW 2000, Australia.
| |
Collapse
|
15
|
Cui H, Deng M, Zhang Y, Yin F, Liu J. Geniposide Increases Unfolded Protein Response-Mediating HRD1 Expression to Accelerate APP Degradation in Primary Cortical Neurons. Neurochem Res 2018; 43:669-680. [DOI: 10.1007/s11064-018-2469-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 12/03/2017] [Accepted: 01/08/2018] [Indexed: 02/20/2023]
|
16
|
Shan M, Yu S, Yan H, Guo S, Xiao W, Wang Z, Zhang L, Ding A, Wu Q, Li SFY. A Review on the Phytochemistry, Pharmacology, Pharmacokinetics and Toxicology of Geniposide, a Natural Product. Molecules 2017; 22:E1689. [PMID: 28994736 PMCID: PMC6151614 DOI: 10.3390/molecules22101689] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 09/27/2017] [Accepted: 10/10/2017] [Indexed: 11/17/2022] Open
Abstract
Iridoid glycosides are natural products occurring widely in many herbal plants. Geniposide (C17H24O10) is a well-known one, present in nearly 40 species belonging to various families, especially the Rubiaceae. Along with this herbal component, dozens of its natural derivatives have also been isolated and characterized by researchers. Furthermore, a large body of pharmacological evidence has proved the various biological activities of geniposide, such as anti-inflammatory, anti-oxidative, anti-diabetic, neuroprotective, hepatoprotective, cholagogic effects and so on. However, there have been some research articles on its toxicity in recent years. Therefore, this review paper aims to provide the researchers with a comprehensive profile of geniposide on its phytochemistry, pharmacology, pharmacokinetics and toxicology in order to highlight some present issues and future perspectives as well as to help us develop and utilize this iridoid glycoside more efficiently and safely.
Collapse
Affiliation(s)
- Mingqiu Shan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sheng Yu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hui Yan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sheng Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Wei Xiao
- National Key Laboratory of Pharmaceutical New Technology for Chinese Medicine, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang 222001, China.
| | - Zhenzhong Wang
- National Key Laboratory of Pharmaceutical New Technology for Chinese Medicine, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang 222001, China.
| | - Li Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Anwei Ding
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Qinan Wu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Sam Fong Yau Li
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore.
| |
Collapse
|