1
|
Zhu J, Guo J, Liu Z, Liu J, Yuan A, Chen H, Qiu J, Dou X, Lu D, Le Y. Salvianolic acid A attenuates non-alcoholic fatty liver disease by regulating the AMPK-IGFBP1 pathway. Chem Biol Interact 2024; 400:111162. [PMID: 39047806 DOI: 10.1016/j.cbi.2024.111162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects approximately a quarter of the population and, to date, there is no approved drug therapy for this condition. Individuals with type 2 diabetes mellitus (T2DM) are at a significantly elevated risk of developing NAFLD, underscoring the urgency of identifying effective NAFLD treatments for T2DM patients. Salvianolic acid A (SAA) is a naturally occurring phenolic acid that is an important component of the water-soluble constituents isolated from the roots of Salvia miltiorrhiza Bunge. SAA has been demonstrated to possess anti-inflammatory and antioxidant stress properties. Nevertheless, its potential in ameliorating diabetes-associated NAFLD has not yet been fully elucidated. In this study, diabetic ApoE-/- mice were employed to establish a NAFLD model via a Western diet. Following this, they were treated with different doses of SAA (10 mg/kg, 20 mg/kg) via gavage. The study demonstrated a marked improvement in liver injury, lipid accumulation, inflammation, and the pro-fibrotic phenotype after the administration of SAA. Additionally, RNA-seq analysis indicated that the primary pathway by which SAA alleviates diabetes-induced NAFLD involves the cascade pathways of lipid metabolism. Furthermore, SAA was found to be effective in the inhibition of lipid accumulation, mitochondrial dysfunction and ferroptosis. A functional enrichment analysis of RNA-seq data revealed that SAA treatment modulates the AMPK pathway and IGFBP-1. Further experimental results demonstrated that SAA is capable of inhibiting lipid accumulation through the activation of the AMPK pathway and IGFBP-1.
Collapse
Affiliation(s)
- Ji Zhu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou, 330106, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhijun Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jing Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Aini Yuan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Hang Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yifei Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
2
|
Yan S, Santoro A, Niphakis MJ, Pinto AM, Jacobs CL, Ahmad R, Suciu RM, Fonslow BR, Herbst-Graham RA, Ngo N, Henry CL, Herbst DM, Saghatelian A, Kahn BB, Rosen ED. Inflammation causes insulin resistance in mice via interferon regulatory factor 3 (IRF3)-mediated reduction in FAHFA levels. Nat Commun 2024; 15:4605. [PMID: 38816388 PMCID: PMC11139994 DOI: 10.1038/s41467-024-48220-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/24/2024] [Indexed: 06/01/2024] Open
Abstract
Obesity-induced inflammation causes metabolic dysfunction, but the mechanisms remain elusive. Here we show that the innate immune transcription factor interferon regulatory factor (IRF3) adversely affects glucose homeostasis through induction of the endogenous FAHFA hydrolase androgen induced gene 1 (AIG1) in adipocytes. Adipocyte-specific knockout of IRF3 protects male mice against high-fat diet-induced insulin resistance, whereas overexpression of IRF3 or AIG1 in adipocytes promotes insulin resistance on a high-fat diet. Furthermore, pharmacological inhibition of AIG1 reversed obesity-induced insulin resistance and restored glucose homeostasis in the setting of adipocyte IRF3 overexpression. We, therefore, identify the adipocyte IRF3/AIG1 axis as a crucial link between obesity-induced inflammation and insulin resistance and suggest an approach for limiting the metabolic dysfunction accompanying obesity.
Collapse
Affiliation(s)
- Shuai Yan
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
| | - Anna Santoro
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
| | - Micah J Niphakis
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Antonio M Pinto
- The Salk Institute for Biological Studies, 10010 N. Torey Pines Rd, La Jolla, CA, 92037-1002, USA
| | - Christopher L Jacobs
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Jasim Mohamad Al Bahar St., Kuwait City, Kuwait
| | - Radu M Suciu
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Bryan R Fonslow
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Rachel A Herbst-Graham
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Nhi Ngo
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Cassandra L Henry
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Dylan M Herbst
- Lundbeck La Jolla Research Center Inc., 10835 Road To The Cure Dr. #250, San Diego, CA, 92121, USA
| | - Alan Saghatelian
- The Salk Institute for Biological Studies, 10010 N. Torey Pines Rd, La Jolla, CA, 92037-1002, USA
| | - Barbara B Kahn
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA
- Broad Institute of Harvard and MIT, 320 Charles St., Cambridge, MA, 02141, USA
| | - Evan D Rosen
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, 330 Brookline Ave, Boston, MA, 02215, USA.
- Harvard Medical School, 25 Shattuck St, Boston, MA, 02130, USA.
- Broad Institute of Harvard and MIT, 320 Charles St., Cambridge, MA, 02141, USA.
| |
Collapse
|
3
|
Zhou Y, Xu B. New insights into anti-diabetes effects and molecular mechanisms of dietary saponins. Crit Rev Food Sci Nutr 2023; 63:12372-12397. [PMID: 35866515 DOI: 10.1080/10408398.2022.2101425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Diabetes mellitus (DM) is a long-term metabolic disorder that manifests as chronic hyperglycemia and impaired insulin, bringing a heavy load on the global health care system. Considering the inevitable side effects of conventional anti-diabetic drugs, saponins-rich natural products exert promising therapeutic properties to serve as safer and more cost-effective alternatives for DM management. Herein, this review systematically summarized the research progress on the anti-diabetic properties of dietary saponins and their underlying molecular mechanisms in the past 20 years. Dietary saponins possessed the multidirectional anti-diabetic capabilities by concurrent regulation of various signaling pathways, such as IRS-1/PI3K/Akt, AMPK, Nrf2/ARE, NF-κB-NLRP3, SREBP-1c, and PPARγ, in liver, pancreas, gut, and skeletal muscle. However, the industrialization and commercialization of dietary saponin-based drugs are confronted with a significant challenge due to the low bioavailability and lack of the standardization. Hence, in-depth evaluations in pharmacological profile, function-structure interaction, drug-signal pathway interrelation are essential for developing dietary saponins-based anti-diabetic treatments in the future.
Collapse
Affiliation(s)
- Yifan Zhou
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, Guangdong, China
- Department of Food Science and Technology, National University of Singapore, Singapore, Singapore
| | - Baojun Xu
- Food Science and Technology Program, BNU-HKBU United International College, Zhuhai, Guangdong, China
| |
Collapse
|
4
|
Mohan J, Ghazi T, Mazibuko MS, Chuturgoon AA. Antiretrovirals Promote Insulin Resistance in HepG2 Liver Cells through miRNA Regulation and Transcriptional Activation of the NLRP3 Inflammasome. Int J Mol Sci 2023; 24:ijms24076267. [PMID: 37047241 PMCID: PMC10094183 DOI: 10.3390/ijms24076267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023] Open
Abstract
Metabolic syndrome (MetS) is a non-communicable disease characterized by a cluster of metabolic irregularities. Alarmingly, the prevalence of MetS in people living with Human Immunodeficiency Virus (HIV) and antiretroviral (ARV) usage is increasing rapidly. Insulin resistance is a common characteristic of MetS that leads to the development of Type 2 diabetes mellitus (T2DM). The progression of insulin resistance is strongly linked to inflammasome activation. This study aimed to draw links between the combinational use of Tenofovir disoproxil fumarate (TDF), Lamivudine (3TC), and Dolutegravir (DTG), and inflammasome activation and subsequent promotion of insulin resistance following a 120 h treatment period in HepG2 liver in vitro cell model. Furthermore, we assess microRNA (miR-128a) expression as a negative regulator of the IRS1/AKT signaling pathway. The relative expression of phosphorylated IRS1 was determined by Western blot. Transcript levels of NLRP3, IL-1β, JNK, IRS1, AKT, PI3K, and miR-128a were assessed using quantitative PCR (qPCR). Caspase-1 activity was measured using luminometry. Following exposure to ARVs for 120 h, NLRP3 mRNA expression (p = 0.0500) and caspase-1 activity (p < 0.0001) significantly increased. This was followed by a significant elevation in IL-1β in mRNA expression (p = 0.0015). Additionally, JNK expression (p = 0.0093) was upregulated with coinciding increases in p-IRS1 protein expression (p < 0.0001) and decreased IRS1 mRNA expression (p = 0.0004). Consequently, decreased AKT (p = 0.0005) and PI3K expressions (p = 0.0007) were observed. Interestingly miR-128a expression was significantly upregulated. The results indicate that combinational use of ARVs upregulates inflammasome activation and promotes insulin resistance through dysregulation of the IRS1/PI3K/AKT insulin signaling pathway.
Collapse
|
5
|
Zhang M, Chen Y, Chen R, Wen Y, Huang Q, Liu Y, Zhao C. Research status of the effects of natural oligosaccharides on glucose metabolism. EFOOD 2022. [DOI: 10.1002/efd2.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Minjiao Zhang
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Yaobin Chen
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Ruoxin Chen
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Yuxi Wen
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Department of Analytical and Food Chemistry, Nutrition and Bromatology Group, Faculty of Sciences Universidade de Vigo Ourense Spain
| | - Qihui Huang
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Department of Analytical and Food Chemistry, Nutrition and Bromatology Group, Faculty of Sciences Universidade de Vigo Ourense Spain
| | - Yuanyuan Liu
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
6
|
DeMarsilis A, Reddy N, Boutari C, Filippaios A, Sternthal E, Katsiki N, Mantzoros C. Pharmacotherapy of type 2 diabetes: An update and future directions. Metabolism 2022; 137:155332. [PMID: 36240884 DOI: 10.1016/j.metabol.2022.155332] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
Abstract
Type 2 diabetes (T2D) is a widely prevalent disease with substantial economic and social impact for which multiple conventional and novel pharmacotherapies are currently available; however, the landscape of T2D treatment is constantly changing as new therapies emerge and the understanding of currently available agents deepens. This review aims to provide an updated summary of the pharmacotherapeutic approach to T2D. Each class of agents is presented by mechanism of action, details of administration, side effect profile, cost, and use in certain populations including heart failure, non-alcoholic fatty liver disease, obesity, chronic kidney disease, and older individuals. We also review targets of novel therapeutic T2D agent development. Finally, we outline an up-to-date treatment approach that starts with identification of an individualized goal for glycemic control then selection, initiation, and further intensification of a personalized therapeutic plan for T2D.
Collapse
Affiliation(s)
- Antea DeMarsilis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Niyoti Reddy
- Department of Medicine, School of Medicine, Boston University, Boston, USA
| | - Chrysoula Boutari
- Second Propedeutic Department of Internal Medicine, Hippocration Hospital, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Andreas Filippaios
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Elliot Sternthal
- Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Sindos, Greece; School of Medicine, European University Cyprus, Nicosia, Cyprus.
| | - Christos Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA; Section of Endocrinology, VA Boston Healthcare System, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Han S, You L, Hu Y, Wei S, Liu T, Cho JY, Hu W. Ginsenoside F2 enhances glucose metabolism by modulating insulin signal transduction in human hepatocarcinoma cells. J Ginseng Res 2022; 47:420-428. [DOI: 10.1016/j.jgr.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/04/2022] [Accepted: 10/11/2022] [Indexed: 03/06/2023] Open
|
8
|
Xia H, Scholtes C, Dufour CR, Ouellet C, Ghahremani M, Giguère V. Insulin action and resistance are dependent on a GSK3β-FBXW7-ERRα transcriptional axis. Nat Commun 2022; 13:2105. [PMID: 35440636 PMCID: PMC9019090 DOI: 10.1038/s41467-022-29722-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 03/30/2022] [Indexed: 12/15/2022] Open
Abstract
Insulin resistance, a harbinger of the metabolic syndrome, is a state of compromised hormonal response resulting from the dysregulation of a wide range of insulin-controlled cellular processes. However, how insulin affects cellular energy metabolism via long-term transcriptional regulation and whether boosting mitochondrial function alleviates insulin resistance remains to be elucidated. Herein we reveal that insulin directly enhances the activity of the nuclear receptor ERRα via a GSK3β/FBXW7 signaling axis. Liver-specific deletion of GSK3β or FBXW7 and mice harboring mutations of ERRα phosphosites (ERRα3SA) co-targeted by GSK3β/FBXW7 result in accumulated ERRα proteins that no longer respond to fluctuating insulin levels. ERRα3SA mice display reprogrammed liver and muscle transcriptomes, resulting in compromised energy homeostasis and reduced insulin sensitivity despite improved mitochondrial function. This crossroad of insulin signaling and transcriptional control by a nuclear receptor offers a framework to better understand the complex cellular processes contributing to the development of insulin resistance.
Collapse
Affiliation(s)
- Hui Xia
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Charlotte Scholtes
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Catherine R Dufour
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Carlo Ouellet
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Majid Ghahremani
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada
| | - Vincent Giguère
- Rosalind and Morris Goodman Cancer Research Institute, McGill University, Montréal, QC, H3A 1A3, Canada.
- Department of Biochemistry, Faculty of Medicine and Health Sciences, McGill University, Montréal, QC, H3G 1Y6, Canada.
| |
Collapse
|
9
|
Chen YH, Chen QQ, Wang CL. Treatment and five-year follow-up of type A insulin resistance syndrome: A case report. World J Clin Cases 2022; 10:2522-2528. [PMID: 35434067 PMCID: PMC8968608 DOI: 10.12998/wjcc.v10.i8.2522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/02/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Type A insulin resistance syndrome (TAIRS) is a rare disorder characterized by severe insulin resistance due to defects in insulin receptor signaling. No specific drugs are available for the treatment of TAIRS. We report a case of TAIRS successfully treated with pioglitazone and flutamide for 5 years.
CASE SUMMARY We present the rare case of a female patient aged 11 years and 9 mo with type A insulin resistance and an INSR heterozygous mutation (c.3614C>T), who was treated with a combination of pioglitazone and flutamide. This treatment regimen reduced hemoglobin A1c, fasting insulin and androgen levels.
CONCLUSION Pioglitazone attenuated insulin resistance in this patient with TAIRS, and flutamide ameliorated masculinization.
Collapse
Affiliation(s)
- Yong-Hua Chen
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Qing-Qing Chen
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| | - Chun-Lin Wang
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, Zhejiang Province, China
| |
Collapse
|
10
|
De Sousa RAL, Improta-Caria AC. Regulation of microRNAs in Alzheimer´s disease, type 2 diabetes, and aerobic exercise training. Metab Brain Dis 2022; 37:559-580. [PMID: 35075500 DOI: 10.1007/s11011-022-00903-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 01/03/2022] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common type of dementia. The evolution and aggregation of amyloid beta (β) oligomers is linked to insulin resistance in AD, which is also the major characteristic of type 2 diabetes (T2D). Being physically inactive can contribute to the development of AD and/or T2D. Aerobic exercise training (AET), a type of physical exercise, can be useful in preventing or treating the negative outcomes of AD and T2D. AD, T2D and AET can regulate the expression of microRNAs (miRNAs). Here, we review some of the changes in miRNAs expression regulated by AET, AD and T2D. MiRNAs play an important role in the gene regulation of key signaling pathways in both pathologies, AD and T2D. MiRNA dysregulation is evident in AD and has been associated with several neuropathological alterations, such as the development of a reactive gliosis. Expression of miRNAs are associated with many pathophysiological mechanisms involved in T2D like insulin synthesis, insulin resistance, glucose intolerance, hyperglycemia, intracellular signaling, and lipid profile. AET regulates miRNAs levels. We identified 5 miRNAs (miR-21, miR-29a/b, miR-103, miR-107, and miR-195) that regulate gene expression and are modulated by AET on AD and T2D. The identified miRNAs are potential targets to treat the symptoms of AD and T2D. Thus, AET is a non-pharmacological tool that can be used to prevent and fight the negative outcomes in AD and T2D.
Collapse
Affiliation(s)
- Ricardo Augusto Leoni De Sousa
- Programa Multicêntrico de Pós-Graduação Em Ciências Fisiológicas- Sociedade Brasileira de Fisiologia (SBFis), Universidade Federal Dos Vales Do Jequitinhonha E Mucuri (UFVJM), Campus JK, Rodovia MGT 367, Km 583, Alto da Jacuba, nº 5000, Diamantina, Minas Gerais, CEP 39100-000, Brazil.
| | - Alex Cleber Improta-Caria
- Post-Graduate Program in Medicine and Health, Faculty of Medicine, Federal University of Bahia, Bahia, Brazil
| |
Collapse
|
11
|
Liao W, Xu N, Zhang H, Liao W, Wang Y, Wang S, Zhang S, Jiang Y, Xie W, Zhang Y. Persistent high glucose induced EPB41L4A-AS1 inhibits glucose uptake via GCN5 mediating crotonylation and acetylation of histones and non-histones. Clin Transl Med 2022; 12:e699. [PMID: 35184403 PMCID: PMC8858623 DOI: 10.1002/ctm2.699] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 12/12/2021] [Accepted: 12/20/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Persistent hyperglycemia decreases the sensitivity of insulin-sensitive organs to insulin, owing to which cells fail to take up and utilize glucose, which exacerbates the progression of type 2 diabetes mellitus (T2DM). lncRNAs' abnormal expression is reported to be associated with the progression of diabetes and plays a significant role in glucose metabolism. Herein, we study the detailed mechanism underlying the functions of lncRNA EPB41L4A-AS1in T2DM. METHODS Data from GEO datasets were used to analyze the expression of EPB41L4A-AS1 between insulin resistance or type 2 diabetes patients and the healthy people. Gene expression was evaluated by qRT-PCR and western blotting. Glucose uptake was measured by Glucose Uptake Fluorometric Assay Kit. Glucose tolerance of mice was detected by Intraperitoneal glucose tolerance tests. Cell viability was assessed by CCK-8 assay. The interaction between EPB41L4A-AS1 and GCN5 was explored by RNA immunoprecipitation, RNA pull-down and RNA-FISH combined immunofluorescence. Oxygen consumption rate was tested by Seahorse XF Mito Stress Test. RESULTS EPB41L4A-AS1 was abnormally increased in the liver of patients with T2DM and upregulated in the muscle cells of patients with insulin resistance and in T2DM cell models. The upregulation was associated with increased TP53 expression and reduced glucose uptake. Mechanistically, through interaction with GCN5, EPB41L4A-AS1 regulated histone H3K27 crotonylation in the GLUT4 promoter region and nonhistone PGC1β acetylation, which inhibited GLUT4 transcription and suppressed glucose uptake by muscle cells. In contrast, EPB41L4A-AS1 binding to GCN5 enhanced H3K27 and H3K14 acetylation in the TXNIP promoter region, which activated transcription by promoting the recruitment of the transcriptional activator MLXIP. This enhanced GLUT4/2 endocytosis and further suppressed glucose uptake. CONCLUSION Our study first showed that the EPB41L4A-AS1/GCN5 complex repressed glucose uptake via targeting GLUT4/2 and TXNIP by regulating histone and nonhistone acetylation or crotonylation. Since a weaker glucose uptake ability is one of the major clinical features of T2DM, the inhibition of EPB41L4A-AS1 expression seems to be a potentially effective strategy for drug development in T2DM treatment.
Collapse
Affiliation(s)
- Weijie Liao
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- School of Life SciencesTsinghua UniversityBeijingP. R. China
| | - Naihan Xu
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Open FIESTA CenterTsinghua UniversityShenzhenP. R. China
| | - Haowei Zhang
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- School of Life SciencesTsinghua UniversityBeijingP. R. China
| | - Weifang Liao
- College of life science and technologyWuhan Polytechnic UniversityWuhanP. R. China
| | - Yanzhi Wang
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- School of Life SciencesTsinghua UniversityBeijingP. R. China
| | - Songmao Wang
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- School of Life SciencesTsinghua UniversityBeijingP. R. China
| | - Shikuan Zhang
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- School of Life SciencesTsinghua UniversityBeijingP. R. China
| | - Yuyang Jiang
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
| | - Weidong Xie
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Open FIESTA CenterTsinghua UniversityShenzhenP. R. China
| | - Yaou Zhang
- State Key Laboratory of Chemical OncogenomicsTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Key Lab in Healthy Science and TechnologyDivision of Life ScienceTsinghua Shenzhen International Graduate SchoolTsinghua UniversityShenzhenP. R. China
- Open FIESTA CenterTsinghua UniversityShenzhenP. R. China
| |
Collapse
|
12
|
Exploring New Drug Targets for Type 2 Diabetes: Success, Challenges and Opportunities. Biomedicines 2022; 10:biomedicines10020331. [PMID: 35203540 PMCID: PMC8869656 DOI: 10.3390/biomedicines10020331] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 01/02/2023] Open
Abstract
There are substantial shortcomings in the drugs currently available for treatment of type 2 diabetes mellitus. The global diabetic crisis has not abated despite the introduction of new types of drugs and targets. Persistent unaddressed patient needs remain a significant factor in the quest for new leads in routine studies. Drug discovery methods in this area have followed developments in the market, contributing to a recent rise in the number of molecules. Nevertheless, troubling developments and fresh challenges are still evident. Recently, metformin, the most widely used first-line drug for diabetes, was found to contain a carcinogenic contaminant known as N-nitroso dimethylamine (NDMA). Therefore, purity and toxicity are also a big challenge for drug discovery and development. Moreover, newer drug classes against SGLT-2 illustrate both progress and difficulties. The same was true previously in the case of glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors. Furthermore, researchers must study the importance of mechanistic characteristics of novel compounds, as well as exposure-related hazardous aspects of current and newly identified protein targets, in order to identify new pharmacological molecules with improved selectivity and specificity.
Collapse
|
13
|
Distribution of TyG index and homeostasis model assessment insulin resistance for the evaluation of insulin sensitivity on late adolescence in Mexicans. NUTR HOSP 2022; 39:1349-1356. [DOI: 10.20960/nh.04120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
14
|
Zeng X, Li Z, Zhu C, Xu L, Sun Y, Han S. Research progress of nanocarriers for gene therapy targeting abnormal glucose and lipid metabolism in tumors. Drug Deliv 2021; 28:2329-2347. [PMID: 34730054 PMCID: PMC8567922 DOI: 10.1080/10717544.2021.1995081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In recent years, the incidence of various types of tumors has gradually increased, and it has also been found that there is a certain correlation between abnormal glucose and lipid metabolism and tumors. Glycolipid metabolism can promote tumor progression through multiple pathways, and the expression of related genes also directly or indirectly affects tumor metabolism, metastasis, invasion, and apoptosis. There has been much research on targeted drug delivery systems designed for abnormal glucose and lipid metabolism due to their accuracy and efficiency when used for tumor therapy. In addition, gene mutations have become an important factor in tumorigenesis. For this reason, gene therapy consisting of drugs designed for certain specifically expressed genes have been transfected into target cells to express or silence the corresponding proteins. Targeted gene drug vectors that achieve their corresponding therapeutic purposes are also rapidly developing. The genes related to glucose and lipid metabolism are considered as the target, and a corresponding gene drug carrier is constructed to influence and interfere with the expression of related genes, so as to block the tumorigenesis process and inhibit tumor growth. Designing drugs that target genes related to glucose and lipid metabolism within tumors is considered to be a promising strategy for the treatment of tumor diseases. This article summarizes the chemical drugs/gene drug delivery systems and the corresponding methods used in recent years for the treatment of abnormal glucose and lipid metabolism of tumors, and provides a theoretical basis for the development of glucolipid metabolism related therapeutic methods.
Collapse
Affiliation(s)
- Xianhu Zeng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Chunrong Zhu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Lisa Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Shangcong Han
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Zhang Y, Wang D, Lv B, Hou X, Liu Q, Liao C, Xu R, Zhang Y, Xu F, Zhang P. Oleic Acid and Insulin as Key Characteristics of T2D Promote Colorectal Cancer Deterioration in Xenograft Mice Revealed by Functional Metabolomics. Front Oncol 2021; 11:685059. [PMID: 34434893 PMCID: PMC8381473 DOI: 10.3389/fonc.2021.685059] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers with high mortality worldwide. Type 2 diabetes mellitus (T2D), known as a risk factor of CRC, can promote the deterioration of CRC, but the underlying mechanism is elusive. In this study, we aimed to reveal the relationship between CRC and T2D from the perspective of small-molecule metabolism. First, a list of common dysregulated metabolites in CRC and T2D was obtained by retrieving existing metabolomics publications. Among these metabolites, oleic acid (OA) was found to be able to promote the proliferation and migration of colon carcinoma cell HCT116. Further experiments proved that insulin could significantly strengthen this promotion and showed a synergistic effect with OA. Mechanism study found that OA and insulin acted synergistically through the extracellular signal-regulated kinase (ERK)1/2/c-Myc/cyclin D1 pathway. In addition, the combination of ERK1/2 inhibitor SCH772984 and cyclin-dependent kinase (CDK)4/6 inhibitor palbociclib showed a remarkable inhibitory effect on tumor growth in vivo. Taken together, the current study found that OA plays an important role in CRC development by using a functional metabolomics approach. More importantly, insulin and OA were confirmed to synergistically promote the deterioration of CRC in vitro and in vivo via ERK1/2/c-Myc/cyclin D1 pathway. Our findings may shed light on CRC treatment among the T2D population.
Collapse
Affiliation(s)
- Ying Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Di Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Bo Lv
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Xiaoying Hou
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Qiwei Liu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Chuyao Liao
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Ruijie Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Yuxin Zhang
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Fengguo Xu
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Pei Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicine, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
16
|
Insulin Signal Transduction Perturbations in Insulin Resistance. Int J Mol Sci 2021; 22:ijms22168590. [PMID: 34445300 PMCID: PMC8395322 DOI: 10.3390/ijms22168590] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus is a widespread medical condition, characterized by high blood glucose and inadequate insulin action, which leads to insulin resistance. Insulin resistance in insulin-responsive tissues precedes the onset of pancreatic β-cell dysfunction. Multiple molecular and pathophysiological mechanisms are involved in insulin resistance. Insulin resistance is a consequence of a complex combination of metabolic disorders, lipotoxicity, glucotoxicity, and inflammation. There is ample evidence linking different mechanistic approaches as the cause of insulin resistance, but no central mechanism is yet described as an underlying reason behind this condition. This review combines and interlinks the defects in the insulin signal transduction pathway of the insulin resistance state with special emphasis on the AGE-RAGE-NF-κB axis. Here, we describe important factors that play a crucial role in the pathogenesis of insulin resistance to provide directionality for the events. The interplay of inflammation and oxidative stress that leads to β-cell decline through the IAPP-RAGE induced β-cell toxicity is also addressed. Overall, by generating a comprehensive overview of the plethora of mechanisms involved in insulin resistance, we focus on the establishment of unifying mechanisms to provide new insights for the future interventions of type 2 diabetes mellitus.
Collapse
|
17
|
Alshahrani S, Anwer T, Alam MF, Ahmed RA, Khan G, Sivakumar SM, Shoaib A, Alam P, Azam F. Effect of thymoquinone on high fat diet and STZ-induced experimental type 2 diabetes: A mechanistic insight by in vivo and in silico studies. J Food Biochem 2021; 45:e13807. [PMID: 34152002 DOI: 10.1111/jfbc.13807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/07/2021] [Accepted: 05/10/2021] [Indexed: 12/24/2022]
Abstract
The aim was to investigate whether thymoquinone (TQ) attenuates hyperglycemia-induced insulin resistance in experimental type 2 diabetes. Type 2 diabetes mellitus (T2DM) was induced by injection of streptozotocin (STZ, 40 mg/kg) in high fat diet (HFD) rats. The levels of glucose, insulin, area under curve (AUC) of glucose, lipid profile parameters, homeostasis model assessment of insulin resistance (HOMA-IR), peroxisome proliferator-activated receptor-γ (PPARγ), and dipeptidyl peptidase peptidase-IV (DPP-IV) were evaluated in HFD + STZ-induced type 2 diabetic rats. TQ treatment significantly reduced elevated levels of glucose, AUC of glucose, insulin, and DPP-IV in diabetic-treated groups. In addition, TQ treatment significantly reduced high levels of triglycerides (TG) and cholesterols (total, low-density and very low-density lipoprotein) accompanied by significant augmentation in high-density lipoprotein (HDL) levels in diabetic-treated groups. However, TQ treatment significantly improved insulin sensitivity in diabetic-treated groups, which was confirmed by increased level of PPARγ and decreased level of HOMA-IR. Molecular docking of TQ exhibited substantial binding affinity with PPARγ and DPP-IV target proteins, which is supported by in vivo results. These results demonstrate that TQ attenuates hyperglycemia-induced insulin resistance by counteracting hyperinsulinemia, improving lipid profile, insulin sensitivity, and inhibiting DPP-IV. PRACTICAL APPLICATIONS: T2DM results in relentless hyperglycemia which eventually progress to a state of insulin resistance. TQ is an active principle compound found in Nigella sative seed, having myriad of traditional medicinal values. Administration of TQ significantly prevented hyperglycemia, hyperinsulinemia, hyperlipidemia, insulin resistance, and inhibited DPP-IV in experimental type 2 diabetes. The in vivo results are also supported by molecular docking study of PPARγ and DPP-IV target proteins. Thus, we hypothesize that TQ can be used as an alternative natural drug in the management of hyperglycemia-induced insulin resistance in T2DM.
Collapse
Affiliation(s)
- Saeed Alshahrani
- Pharmacology & Toxicology Department, Pharmacy College, Jazan University, Gizan, Saudi Arabia
| | - Tarique Anwer
- Pharmacology & Toxicology Department, Pharmacy College, Jazan University, Gizan, Saudi Arabia
| | - Mohammad Firoz Alam
- Pharmacology & Toxicology Department, Pharmacy College, Jazan University, Gizan, Saudi Arabia
| | - Rayan A Ahmed
- Pharmacology & Toxicology Department, Pharmacy College, Jazan University, Gizan, Saudi Arabia
| | - Gyas Khan
- Pharmacology & Toxicology Department, Pharmacy College, Jazan University, Gizan, Saudi Arabia
| | | | - Ambreen Shoaib
- Clinical Pharmacy Department, Pharmacy College, Jazan University, Gizan, Saudi Arabia
| | - Prawez Alam
- Pharmacognosy Department, Pharmacy College, Prince Sattam bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Faizul Azam
- Pharmaceutical Chemistry & Pharmacognosy Department, Unaizah College of Pharmacy, Qassim University, Unaizah, Saudi Arabia
| |
Collapse
|
18
|
de Oliveira WQ, Neri-Numa IA, Arruda HS, Lopes AT, Pelissari FM, Barros FFC, Pastore GM. Special emphasis on the therapeutic potential of microparticles with antidiabetic effect: Trends and possible applications. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.02.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
19
|
Transition of Hypertriglyceridemic-Waist Phenotypes and the Risk of Type 2 Diabetes Mellitus among Middle-Aged and Older Chinese: A National Cohort Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18073664. [PMID: 33915915 PMCID: PMC8037185 DOI: 10.3390/ijerph18073664] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/23/2021] [Accepted: 03/28/2021] [Indexed: 10/26/2022]
Abstract
The rapid economic growth and nutritional changes in China have brought an increased burden of type 2 diabetes mellitus (T2DM). This study aimed to assess the effects of hypertriglyceridemic-waist (HTW) and its dynamic transitions on incident T2DM among middle-aged and older Chinese. Data were extracted from the China Health and Retirement Longitudinal Study (CHARLS). Participants were classified into three HTW phenotypes, namely NTNW (normal triglyceride (TG) and waist circumference (WC)), NTEW/ETNW (normal TG and enlarged WC, or elevated TG and normal WC) and ETEW (elevated TG and enlarged WC). Multivariable Cox frailty models were used to assess the associations of HTW phenotypes and their transitions over time with the risk of T2DM. A total of 7397 subjects without T2DM were included, of which 849 developed T2DM during 2011-2018. Compared with individuals with NTNW, people in the NTEW/ETNW group and ETEW group were at a significantly higher risk of T2DM (HRNTEW/ETNW = 1.28, 95% CI: 1.06-1.54 and HRETEW = 1.61, 95% CI: 1.26-2.06). For subjects with NTNW at baseline, the risk of developing T2DM increased by 38% and 83% if their metabolic status changed to NTEW/ETNW and ETEW, respectively. For subjects with NTEW/ETNW, the risk of T2DM decreased by 33% when their metabolic status changed to normal (NTNW); but the risk increased by 49% if the status became more serious (ETEW). NTEW/ETNW, ETEW and their transitions to adverse states were risk factors for T2DM.
Collapse
|
20
|
Behl T, Kaur I, Sehgal A, Singh S, Zengin G, Negrut N, Nistor-Cseppento DC, Pavel FM, Corb Aron RA, Bungau S. Exploring the Genetic Conception of Obesity via the Dual Role of FoxO. Int J Mol Sci 2021. [DOI: https://doi.org/10.3390/ijms22063179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Obesity or overweight are not superficial problems, constituting a pressing issue. The obesity index has almost tripled since 1975, which is an alarming state. Most of the individuals are currently becoming overweight or have inappropriate body mass index (BMI) conditions. Obesity is characterized by increased fat accumulation and thus poses a higher health risk. There is increased size and volume of fat cells in the body, which usually accounts for obesity. Many investigations have been carried out in this area, such as behavioral improvements, dietary changes, chemical involvements, etc., but presently no such goals are established to manage these health concerns. Based on previous literature reports and our interpretation, the current review indicates the involvement of various transcriptional and transporter functions in modifying the above-mentioned health conditions. Various transcriptional factors such as Forkhead box O1 (FoxO1) impart a significant effect on the physiology and pathology of metabolic dysfunction such as obesity. FoxO1 plays a dual role whether in the progression or suppression of metabolic processes depending on its targets. Thus, in the current study, will be discussed the dual role of FoxO1 in metabolic conditions (such as obesity), also summarizing the role of various other transcriptional factors involved in obesity.
Collapse
|
21
|
Behl T, Kaur I, Sehgal A, Singh S, Zengin G, Negrut N, Nistor-Cseppento DC, Pavel FM, Corb Aron RA, Bungau S. Exploring the Genetic Conception of Obesity via the Dual Role of FoxO. Int J Mol Sci 2021; 22:ijms22063179. [PMID: 33804729 PMCID: PMC8003860 DOI: 10.3390/ijms22063179] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity or overweight are not superficial problems, constituting a pressing issue. The obesity index has almost tripled since 1975, which is an alarming state. Most of the individuals are currently becoming overweight or have inappropriate body mass index (BMI) conditions. Obesity is characterized by increased fat accumulation and thus poses a higher health risk. There is increased size and volume of fat cells in the body, which usually accounts for obesity. Many investigations have been carried out in this area, such as behavioral improvements, dietary changes, chemical involvements, etc., but presently no such goals are established to manage these health concerns. Based on previous literature reports and our interpretation, the current review indicates the involvement of various transcriptional and transporter functions in modifying the above-mentioned health conditions. Various transcriptional factors such as Forkhead box O1 (FoxO1) impart a significant effect on the physiology and pathology of metabolic dysfunction such as obesity. FoxO1 plays a dual role whether in the progression or suppression of metabolic processes depending on its targets. Thus, in the current study, will be discussed the dual role of FoxO1 in metabolic conditions (such as obesity), also summarizing the role of various other transcriptional factors involved in obesity.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| | - Ishnoor Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (I.K.); (A.S.); (S.S.)
| | - Gokhan Zengin
- Department of Biology, Faculty of Science, Selcuk University Campus, Konya 42130, Turkey;
| | - Nicoleta Negrut
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (N.N.); (D.C.N.-C.)
| | - Delia Carmen Nistor-Cseppento
- Department of Psycho-Neuroscience and Recovery, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (N.N.); (D.C.N.-C.)
| | - Flavia Maria Pavel
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (F.M.P.); (R.A.C.A.)
| | - Raluca Anca Corb Aron
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania; (F.M.P.); (R.A.C.A.)
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
- Correspondence: (T.B.); (S.B.); Tel.: +40-726-776-588 (S.B.)
| |
Collapse
|
22
|
Gunder LC, Harvey I, Redd JR, Davis CS, AL-Tamimi A, Brooks SV, Bridges D. Obesity Augments Glucocorticoid-Dependent Muscle Atrophy in Male C57BL/6J Mice. Biomedicines 2020; 8:E420. [PMID: 33076257 PMCID: PMC7602414 DOI: 10.3390/biomedicines8100420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/25/2020] [Accepted: 10/07/2020] [Indexed: 12/22/2022] Open
Abstract
Glucocorticoids promote muscle atrophy by inducing a class of proteins called atrogenes, resulting in reductions in muscle size and strength. In this work, we evaluated whether a mouse model with pre-existing diet-induced obesity had altered glucocorticoid responsiveness. We observed that all animals treated with the synthetic glucocorticoid dexamethasone had reduced strength, but that obesity exacerbated this effect. These changes were concordant with more pronounced reductions in muscle size, particularly in Type II muscle fibers, and potentiated induction of atrogene expression in the obese mice relative to lean mice. Furthermore, we show that the reductions in lean mass do not fully account for the dexamethasone-induced insulin resistance observed in these mice. Together, these data suggest that obesity potentiates glucocorticoid-induced muscle atrophy.
Collapse
Affiliation(s)
- Laura C. Gunder
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; (L.C.G.); (I.H.); (J.R.R.); (A.A.-T.)
| | - Innocence Harvey
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; (L.C.G.); (I.H.); (J.R.R.); (A.A.-T.)
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70803, USA
| | - JeAnna R. Redd
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; (L.C.G.); (I.H.); (J.R.R.); (A.A.-T.)
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Carol S. Davis
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (C.S.D.); (S.V.B.)
| | - Ayat AL-Tamimi
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; (L.C.G.); (I.H.); (J.R.R.); (A.A.-T.)
| | - Susan V. Brooks
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (C.S.D.); (S.V.B.)
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA; (L.C.G.); (I.H.); (J.R.R.); (A.A.-T.)
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA; (C.S.D.); (S.V.B.)
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| |
Collapse
|
23
|
Zhang L, Li X, Zhang N, Yang X, Hou T, Fu W, Yuan F, Wang L, Wen H, Tian Y, Zhang H, Lu X, Zhu WG. WDFY2 Potentiates Hepatic Insulin Sensitivity and Controls Endosomal Localization of the Insulin Receptor and IRS1/2. Diabetes 2020; 69:1887-1902. [PMID: 32641353 DOI: 10.2337/db19-0699] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 06/04/2020] [Indexed: 11/13/2022]
Abstract
Endosomes help activate the hepatic insulin-evoked Akt signaling pathway, but the underlying regulatory mechanisms are unclear. Previous studies have suggested that the endosome-located protein WD repeat and FYVE domain-containing 2 (WDFY2) might be involved in metabolic disorders, such as diabetes. Here, we generated Wdfy2 knockout (KO) mice and assessed the metabolic consequences. These KO mice exhibited systemic insulin resistance, with increased gluconeogenesis and suppressed glycogen accumulation in the liver. Mechanistically, we found that the insulin-stimulated activation of Akt2 and its substrates FoxO1 and GSK-3β is attenuated in the Wdfy2 KO liver and H2.35 hepatocytes, suggesting that WDFY2 acts as an important regulator of hepatic Akt2 signaling. We further found that WDFY2 interacts with the insulin receptor (INSR) via its WD1-4 domain and localizes the INSR to endosomes after insulin stimulation. This process ensures that the downstream insulin receptor substrates 1 and 2 (IRS1/2) can be recruited to the endosomal INSR. IRS1/2-INSR binding promotes IRS1/2 phosphorylation and subsequent activation, initiating downstream Akt2 signaling in the liver. Interestingly, adeno-associated viral WDFY2 delivery ameliorated metabolic defects in db/db mice. These findings demonstrate that WDFY2 activates insulin-evoked Akt2 signaling by controlling endosomal localization of the INSR and IRS1/2 in hepatocytes. This pathway might constitute a new potential target for diabetes prevention or treatment.
Collapse
Affiliation(s)
- Luyao Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xue Li
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Nan Zhang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xin Yang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Tianyun Hou
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| | - Wan Fu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Fengjie Yuan
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lina Wang
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - He Wen
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
| | - Yuan Tian
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Hongquan Zhang
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xifeng Lu
- Department of Physiology, Shenzhen University School of Medicine, Shenzhen, China
| | - Wei-Guo Zhu
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
- Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University School of Medicine, Shenzhen, China
- Shenzhen Bay Laboratory, Shenzhen, China
| |
Collapse
|
24
|
Luo Z, Xu W, Zhang Y, Di L, Shan J. A review of saponin intervention in metabolic syndrome suggests further study on intestinal microbiota. Pharmacol Res 2020; 160:105088. [PMID: 32683035 DOI: 10.1016/j.phrs.2020.105088] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 06/13/2020] [Accepted: 07/14/2020] [Indexed: 12/16/2022]
Abstract
Metabolic syndrome (MetS) is a series of symptoms including insulin resistance, obesity, dyslipidemia, elevated fasting blood glucose levels, and hepatic steatosis. As a key criterion in MetS, the onset of insulin resistance is related to abnormal levels of circulating free fatty acids and adipokines. It has been discovered in recent years that metabolites and pathogen-associated molecular patterns of intestinal/gut microbiota are also important factors that cause insulin resistance and MetS. Saponins are the main components of many botanicals and traditional Chinese medicines (TCMs), such as ginseng, platycodon, licorice, and alfalfa. They have poor bioavailability, but can be transformed into secondary glycosides and aglycones by intestinal microbiota, further being absorbed. Based on in vivo and in vitro data, we found that saponins and their secondary metabolites have a preventive effect on MetS, and the effective targets are distributed in the intestine and other organs in human body. Intestinal targets involve pancreatic lipase, dietary cholesterol, and intestinal microbiota. Other targets include central appetite, nuclear receptors such as PPAR and LXR, AMPK signaling pathway and adipokines levels, etc. In view of the poor bioavailability of saponins, it is inferred that targets for prototype-saponins to interfere with MetS is mainly located in the intestine, and the activation of other targets may be related to secondary glycosides and aglycones transformed from saponins by intestinal flora. We suggest that the role of intestinal microbiota in saponin intervention in MetS should be further investigated.
Collapse
Affiliation(s)
- Zichen Luo
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weichen Xu
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ying Zhang
- Genome Center of UC Davis, NIH West Coast Metabolomics Center, Davis, CA, 95616, USA
| | - Liuqing Di
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jinjun Shan
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing 210023, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing 210023, China; Jiangsu Engineering Research Center for Efficient Delivery System of TCM, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
25
|
Biondo LA, Teixeira AAS, de O. S. Ferreira KC, Neto JCR. Pharmacological Strategies for Insulin Sensitivity in Obesity and Cancer: Thiazolidinediones and Metformin. Curr Pharm Des 2020; 26:932-945. [DOI: 10.2174/1381612826666200122124116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/21/2019] [Indexed: 12/19/2022]
Abstract
Background:
Chronic diseases, such as obesity and cancer, have high prevalence rates. Both diseases
have hyperinsulinemia, hyperglycemia, high levels of IGF-1 and inflammatory cytokines in common. Therefore,
these can be considered triggers for cancer development and growth. In addition, low-grade inflammation that
modulates the activation of immune cells, cellular metabolism, and production of cytokines and chemokines are
common in obesity, cancer, and insulin resistance. Pharmacological strategies are necessary when a change in
lifestyle does not improve glycemic homeostasis. In this regard, thiazolidinediones (TZD) possess multiple molecular
targets and regulate PPARγ in obesity and cancer related to insulin resistance, while metformin acts
through the AMPK pathway.
Objective:
The aim of this study was to review TZD and metformin as pharmacological treatments for insulin
resistance associated with obesity and cancer.
Conclusions:
Thiazolidinediones restored adiponectin secretion and leptin sensitivity, reduced lipid droplets in
hepatocytes and orexigen peptides in the hypothalamus. In cancer cells, TZD reduced proliferation, production of
reactive oxygen species, and inflammation by acting through the mTOR and NFκB pathways. Metformin has
similar effects, though these are AMPK-dependent. In addition, both drugs can be efficient against certain side
effects caused by chemotherapy.
Collapse
Affiliation(s)
- Luana A. Biondo
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Alexandre A. S. Teixeira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Karen C. de O. S. Ferreira
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Jose C. R. Neto
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
26
|
Aboonabi A, Aboonabi A. Anthocyanins reduce inflammation and improve glucose and lipid metabolism associated with inhibiting nuclear factor-kappaB activation and increasing PPAR-γ gene expression in metabolic syndrome subjects. Free Radic Biol Med 2020; 150:30-39. [PMID: 32061902 DOI: 10.1016/j.freeradbiomed.2020.02.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 01/31/2020] [Accepted: 02/09/2020] [Indexed: 02/06/2023]
Abstract
Anthocyanins exhibit antioxidant and anti-inflammatory activities via a multitude of biochemical mechanisms. However, the signaling pathways involved in the actions of anthocyanins against chronic inflammation are not fully understood. The effects of berry-rich anthocyanin supplements (320 mg/day) for four weeks were examined on features of metabolic syndrome components and the expression of PPAR-γ, Nrf2, and NF-κB dependent genes in MetS and healthy subjects. Total RNA was isolated from whole blood with the PAXgene proprietary blood collection system. Four weeks anthocyanin consumption significantly decreased fasting blood glucose (15.7% vs 3.2%), TG (18.2% vs -1.39%), cholesterol (33.5% vs 1.56%) and LDL (28.4% vs -15.6%) in the MetS compared to Control group (P-value < 0.05, 95% CI). There was a significant up regulation in the expression PPAR-γ gene associated with the lipid and glucose metabolism in MetS subjects which negatively correlated (P-value < 0.01) with the change in the FBG (r = -0.488), Cholesterol (r = -0.496), TG (r = -0.513) and LDL (r = -0.519). Moreover, anthocyanin supplementation decreases serum hs-CRP (-36.3% vs 6.25%) in MetS in compared to Control group (P-value < 0.05). Anthocyanin supplementation also down-regulated the expression of NF-κB dependent genes including TNF-α (-28% and -15%), IL-6 (-16.1% and -13.6%), IL-1A (-21.5% and -12.9%), PCAM-1 (-15% and -17.5%), and COX-2(-26% and -27%) in both MetS and Control group respectively (P-value < 0.05). The study results suggested that berry supplements improved selected features of metabolic syndrome and related cardiovascular risk factors. These benefits may be due to the inhibition of NF-κB dependent gene expression and enhancement of PPAR-γ.
Collapse
Affiliation(s)
- Anahita Aboonabi
- School of Medical Science, Gold Coast Campus, Griffith University, Parklands Drive, Southport, Queensland, 4222, Australia.
| | - Arta Aboonabi
- West Center of Tehran, Payam Noor University, Shahid Bagheri Town, Tehran, Iran.
| |
Collapse
|
27
|
Czech MP. Mechanisms of insulin resistance related to white, beige, and brown adipocytes. Mol Metab 2020; 34:27-42. [PMID: 32180558 PMCID: PMC6997501 DOI: 10.1016/j.molmet.2019.12.014] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/21/2019] [Accepted: 12/23/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The diminished glucose lowering effect of insulin in obesity, called "insulin resistance," is associated with glucose intolerance, type 2 diabetes, and other serious maladies. Many publications on this topic have suggested numerous hypotheses on the molecular and cellular disruptions that contribute to the syndrome. However, significant uncertainty remains on the mechanisms of its initiation and long-term maintenance. SCOPE OF REVIEW To simplify insulin resistance analysis, this review focuses on the unifying concept that adipose tissue is a central regulator of systemic glucose homeostasis by controlling liver and skeletal muscle metabolism. Key aspects of adipose function related to insulin resistance reviewed are: 1) the modes by which specific adipose tissues control hepatic glucose output and systemic glucose disposal, 2) recently acquired understanding of the underlying mechanisms of these modes of regulation, and 3) the steps in these pathways adversely affected by obesity that cause insulin resistance. MAJOR CONCLUSIONS Adipocyte heterogeneity is required to mediate the multiple pathways that control systemic glucose tolerance. White adipocytes specialize in sequestering triglycerides away from the liver, muscle, and other tissues to limit toxicity. In contrast, brown/beige adipocytes are very active in directly taking up glucose in response to β adrenergic signaling and insulin and enhancing energy expenditure. Nonetheless, white, beige, and brown adipocytes all share the common feature of secreting factors and possibly exosomes that act on distant tissues to control glucose homeostasis. Obesity exerts deleterious effects on each of these adipocyte functions to cause insulin resistance.
Collapse
Affiliation(s)
- Michael P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| |
Collapse
|
28
|
|
29
|
Festuccia WT. Regulation of Adipocyte and Macrophage Functions by mTORC1 and 2 in Metabolic Diseases. Mol Nutr Food Res 2020; 65:e1900768. [DOI: 10.1002/mnfr.201900768] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/06/2020] [Indexed: 12/13/2022]
Affiliation(s)
- William T. Festuccia
- Department of Physiology and Biophysics Institute of Biomedical Sciences University of Sao Paulo Sao Paulo 05508000 Brazil
| |
Collapse
|
30
|
Therapeutic potential of ginsenosides on diabetes: From hypoglycemic mechanism to clinical trials. J Funct Foods 2020. [DOI: 10.1016/j.jff.2019.103630] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
31
|
Cao D, Wang W, Li S, Lai W, Huang X, Zhou J, Chen X, Li X. TLR2-Deficiency Promotes Prenatal LPS Exposure-Induced Offspring Hyperlipidemia. Front Physiol 2019; 10:1102. [PMID: 31507457 PMCID: PMC6713936 DOI: 10.3389/fphys.2019.01102] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022] Open
Abstract
Toll-like receptor 2 (TLR2), which recognizes several lipopeptides and transduces inflammatory signaling, promotes the pathogenesis of diet-induced dyslipidemia and obesity. TLR2-deficient mice were shown to have improved insulin sensitivity and reduced diet-induced metabolic syndrome. Previous studies demonstrated that prenatal lipopolysaccharide (LPS) exposure causes dyslipidemia accompanied by increased body weight and insulin resistance in offspring. To determine whether TLRs are involved in this complex abnormal phenotype, we analyzed TLR2 and TLR4 expression levels in adipose tissues from offspring with prenatal LPS-exposure (offspring-pLPS) and compared these levels to those of control offspring with prenatal saline-exposure (offspring-pSaline). TLR2 expression was specifically upregulated in the adipose tissue of offspring-pLPS mice. However, unexpectedly, TLR2-deficient offspring-pLPS mice not only presented with an abnormal phenotype comparable to that of wild-type offspring-pLPS mice but also exhibited significantly more severe hyperlipidemia. Our further analyses revealed a dramatic upregulation of TLR4 expression and overactivation of the TLR4/Myd88 signaling pathway in TLR2-deficient offspring-pLPS adipose tissue. Our finding suggests a compensatory genetic interaction between TLR2 and TLR4 in the context of prenatal inflammatory stimulation, and this interaction likely contributes to the prenatal inflammation-induced hyperlipidemia and lipid overload-induced obesity, thus providing a potential mechanism for the fetal origin of adult metabolic diseases.
Collapse
Affiliation(s)
- Dayan Cao
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Wenjia Wang
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Shuhui Li
- Department of Clinical Biochemistry, College of Pharmacy, Army Medical University, Chongqing, China
| | - Wenjing Lai
- Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaoyong Huang
- Institute of Immunology, PLA, Army Medical University, Chongqing, China
| | - Jianzhi Zhou
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Xin Chen
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, Department of Pharmaceutics, College of Pharmacy, Army Medical University, Chongqing, China
| |
Collapse
|
32
|
Zhu D, Yan Q, Liu J, Wu X, Jiang Z. Can functional oligosaccharides reduce the risk of diabetes mellitus? FASEB J 2019; 33:11655-11667. [PMID: 31415188 DOI: 10.1096/fj.201802802rrr] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Diabetes significantly affects the life quality and length of patients with diabetes, and almost half of the 4 million people who die from diabetes are under the age of 60. Because of the increasing number of patients with diabetes and the side effects of antidiabetic drugs, the search for new dietary supplementation from natural resources, especially functional oligosaccharides, has attracted much attention among scientific researchers. Functional oligosaccharides are potential antidiabetic treatments because of their nondigestible, low-calorie, and probiotic features. The antidiabetic activity of multiple functional oligosaccharides such as fructo-oligosaccharides, galacto-oligosaccharides, and xylo-oligosaccharides has been reviewed in this paper. Molecular mechanisms involved in the antidiabetic activity of oligosaccharides have been systematically discussed from multiple perspectives, including the improvement of pancreas function, α-glucosidase inhibition, the relief of insulin and leptin resistance, anti-inflammatory effects, regulation of gut microbiota and hormones, and the intervention of diabetic risk factors. In addition, the antidiabetic effects of functional oligosaccharides through the complex gut-brain-liver axis are summarized. The concepts addressed in this review have important clinical implications, although more works are needed to confirm the antidiabetic mechanisms of functional oligosaccharides, standardize safe dose levels, and clarify their metabolism in the human body.-Zhu, D., Yan, Q., Liu, J., Wu, X., Jiang, Z. Can functional oligosaccharides reduce the risk of diabetes mellitus?
Collapse
Affiliation(s)
- Di Zhu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qiaojuan Yan
- College of Engineering, China Agricultural University, Beijing, China
| | - Jun Liu
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xia Wu
- College of Engineering, China Agricultural University, Beijing, China
| | - Zhengqiang Jiang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
33
|
Baken KA, Lambrechts N, Remy S, Mustieles V, Rodríguez-Carrillo A, Neophytou CM, Olea N, Schoeters G. A strategy to validate a selection of human effect biomarkers using adverse outcome pathways: Proof of concept for phthalates and reproductive effects. ENVIRONMENTAL RESEARCH 2019; 175:235-256. [PMID: 31146096 DOI: 10.1016/j.envres.2019.05.013] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/09/2019] [Accepted: 05/10/2019] [Indexed: 05/21/2023]
Abstract
Human biomonitoring measures the concentrations of environmental chemicals or their metabolites in body fluids or tissues. Complementing exposure biomarkers with mechanistically based effect biomarkers may further elucidate causal pathways between chemical exposure and adverse health outcomes. We combined information on effect biomarkers previously implemented in human observational studies with mechanisms of action reported in experimental studies and with information from published Adverse Outcome Pathways (AOPs), focusing on adverse reproductive effects of phthalate exposure. Phthalates constitute a group of chemicals that are ubiquitous in consumer products and have been related to a wide range of adverse health effects. As a result of a comprehensive literature search, we present an overview of effect biomarkers for reproductive toxicity that are substantiated by mechanistic information. The activation of several receptors, such as PPARα, PPARγ, and GR, may initiate events leading to impaired male and female fertility as well as other adverse effects of phthalate exposure. Therefore, these receptors appear as promising targets for the development of novel effect biomarkers. The proposed strategy connects the fields of epidemiology and toxicology and may strengthen the weight of evidence in observational studies that link chemical exposures to health outcomes.
Collapse
Affiliation(s)
- Kirsten A Baken
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium.
| | - Nathalie Lambrechts
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium
| | - Sylvie Remy
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium; Department of Epidemiology and Social Medicine, University of Antwerp, Antwerp, Belgium
| | - Vicente Mustieles
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, Granada, Spain; Center for Biomedical Research (CIBM), University of Granada, Granada, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain
| | | | - Christiana M Neophytou
- Department of Biological Sciences, School of Pure and Applied Sciences, University of Cyprus, Nicosia, Cyprus
| | - Nicolas Olea
- Biosanitary Research Institute of Granada (ibs.GRANADA), University Hospitals of Granada, Granada, Spain; Center for Biomedical Research (CIBM), University of Granada, Granada, Spain; Consortium for Biomedical Research in Epidemiology & Public Health (CIBERESP), Spain
| | - Greet Schoeters
- Unit Health, Flemish Institute for Technological Research (VITO NV), Mol, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium; Department of Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
34
|
Effect of Konjac Mannan Oligosaccharides on Glucose Homeostasis via the Improvement of Insulin and Leptin Resistance In Vitro and In Vivo. Nutrients 2019; 11:nu11081705. [PMID: 31344867 PMCID: PMC6723648 DOI: 10.3390/nu11081705] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 07/20/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022] Open
Abstract
Functional oligosaccharides, particularly konjac mannan oligosaccharides (KMOS), can regulate glucose metabolism. However, the molecular mechanisms involved in the hypoglycemic effect of KMOS remain largely unknown. Here, the effect of KMOS supplementation on glucose homeostasis was evaluated in both high-fat diet (HFD)-fed C57BL/6J mice and high-glucosamine-induced HepG2 cells. KMOS supplementation remarkably ameliorated the fasting blood glucose, glucose tolerance, and insulin tolerance of HFD-fed mice. Abnormalities of triglyceride and glycogen metabolism in the liver induced by the HFD were reversed by KMOS supplementation. The insulin signaling pathway was activated by KMOS, with stimulation of GLUT2 membrane translocation and glucose uptake in HepG2 cells via the AMPK pathway. Moreover, KMOS suppressed p-mTOR expression and stimulated the GSK-3β/CREB pathway via the AMPK pathway. KMOS significantly upregulated leptin receptor expression and downregulated PTP1B and SOCS3 levels in the liver and brain, with a decreased serum leptin concentration. Phosphorylation of JAK2 and STAT3 in the liver was activated by KMOS supplementation, while the expressions of Sirt1, Tfam, and Pgc1-α in the brain were elevated. Conclusively, KMOS attenuated HFD-induced glucose metabolism dysfunction through the regulation of insulin resistance and leptin resistance. This finding indicates that KMOS have potential value as an anti-hyperglycemic dietary supplement.
Collapse
|
35
|
Abdulwahab RA, Allaith AAA, Shinwari Z, Alaiya A, Giha HA. Association of TATA box-binding protein-associated factor RNA polymerase I subunit C (TAF1C) with T2DM. Gene 2019; 706:43-51. [PMID: 31039436 DOI: 10.1016/j.gene.2019.04.076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/29/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
Abstract
Proteins differential expression in type 2 diabetes mellitus (T2DM) can be due to etiological factors or pathological changes, such proteins can be utilized as biomarkers. Identification of a marker protein out of thousands became a feasible task during the proteomics era by using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In this study, blood samples were obtained from 80 Bahraini subjects with and without T2DM, a subset was used for proteomic analysis by LC-MS/MS, while all samples were used for ELISA analysis of 3 proteins, TATA-box binding protein-associated factor RNA polymerase-1-C (TAF1C), ceruloplasmin (CERP) and fibronectin (FN). The former 2 proteins were selected from the T2DM-protein-panel identified by LC-MS/MS, and the latter was analyzed for validation of the setting. The main findings of the proteomic analysis are i. Identifications of 62 differentially expressed proteins in T2DM, ii. Upregulation of 71% of the identified proteins. While the ELISA analysis showed that; both TAF1C and FN were significantly increased in T2DM (P0.015 and P0.001, respectively), while CERP was not (P0.088). Logistic regression analysis: i. confirmed the above associations after correction for covariates, ii. Revealed an interaction between age and gender that affect the association of the proteins with T2DM. In conclusion, knowing that TAF1C is a prerequisite in ribosomal biogenesis, our ELISA results are suggestive of increased protein synthesis in T2DM, explaining the observed upregulation of the proteins identified by LC-MSMS. The association between T2DM and TAF1C is a novel finding that might open a new avenue in DM research.
Collapse
Affiliation(s)
- Rabab Asghar Abdulwahab
- Integrated Science Division, College of Health Sciences, University of Bahrain, Manama, Bahrain; Al Jawhara Centre for Molecular Medicine and Inherited Disorders, Arabian Gulf University, Bahrain
| | | | - Zakia Shinwari
- Proteomics Unit, Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre (KFSH&RC), P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Ayodele Alaiya
- Proteomics Unit, Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre (KFSH&RC), P.O. Box 3354, Riyadh 11211, Saudi Arabia
| | - Hayder A Giha
- Department of Biochemistry, College of Medicine and Medical Sciences (CMMS), Arabian Gulf University (AGU), Manama, Bahrain.
| |
Collapse
|
36
|
Gong Y, Liu J, Xue Y, Zhuang Z, Qian S, Zhou W, Li X, Qian J, Ding G, Sun Z. Non-monotonic dose-response effects of arsenic on glucose metabolism. Toxicol Appl Pharmacol 2019; 377:114605. [PMID: 31170414 DOI: 10.1016/j.taap.2019.114605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 05/29/2019] [Accepted: 05/31/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND Inorganic arsenic (iAs) is a widespread environmental toxin. In addition to being a human carcinogen, its effect on diabetes has started to gain recognition recently. Insulin is the key hormone regulating systemic glucose metabolism. The in vivo effect of iAs on insulin sensitivity has not been directly addressed. OBJECTIVES Here we use mouse models to dissect the dose-dependent effects of iAs on glucose metabolism in vivo. METHODS We performed hyperinsulinemic-euglycemic clamp, the gold standard analysis of systemic insulin sensitivity. We also performed dynamic metabolic testings and RNA-seq analysis. RESULTS We found that a low-dose exposure (0.25 ppm iAs in drinking water) caused glucose intolerance in adult male C57BL/6 mice, likely by disrupting glucose-induced insulin secretion without affecting peripheral insulin sensitivity. However, a higher-dose exposure (2.5 ppm iAs) had diminished effects on glucose tolerance despite disrupted pancreatic insulin secretion. Insulin Clamp analysis showed that 2.5 ppm iAs actually enhanced systemic insulin sensitivity by simultaneously enhancing insulin-stimulated glucose uptake in skeletal muscles and improved insulin-mediated suppression of endogenous glucose production. RNA-seq analysis of skeletal muscles revealed that 2.5 ppm iAs regulated expression of many genes involved in the metabolism of fatty acids, pyruvate, and amino acids. CONCLUSION These findings suggest that iAs has opposite glycemic effects on distinct metabolic tissues at different dose thresholds. Such non-monotonic dose-response effects of iAs on glucose tolerance shed light on the complex interactions between iAs and the systemic glucose metabolism, which could potentially help reconcile some of the conflicting results in human epidemiological studies.
Collapse
Affiliation(s)
- Yingyun Gong
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Jidong Liu
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Yanfeng Xue
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Zhong Zhuang
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Sichong Qian
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Wenjun Zhou
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Xin Li
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Justin Qian
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America
| | - Guolian Ding
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America; The International Peace Maternity and Child Health Hospital, Institute of Embryo-Fetal Original Adult Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zheng Sun
- Department of Medicine-Endocrinology, Baylor College of Medicine, Houston, TX, United States of America; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States of America.
| |
Collapse
|
37
|
Xing Y, Zhang J, Wei H, Zhang H, Guan Y, Wang X, Tong X. Reduction of the PI3K/Akt related signaling activities in skeletal muscle tissues involves insulin resistance in intrauterine growth restriction rats with catch-up growth. PLoS One 2019; 14:e0216665. [PMID: 31071176 PMCID: PMC6508867 DOI: 10.1371/journal.pone.0216665] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/25/2019] [Indexed: 12/29/2022] Open
Abstract
Individuals with intrauterine growth retardation (IUGR) are prone to insulin resistance, whereas the underlying molecular mechanisms remain unclear. In this study, we investigated if the PI3K/Akt signaling pathway in skeletal muscle tissues involves insulin resistance in IUGR offsprings, particularly ones with catch-up growth. An IUGR rat model was established by feeding rats an isocaloric (30.50 Kcal/g) diet containing 8% protein (low-protein diet) from day 1 of pregnancy until the birth of their pups. Glucometabolic parameters were measured and compared. Quantitative PCR and western blot were performed to assess mRNA and protein expression changes of the PI3K/Akt related signals in skeletal muscle tissues. Pearson analysis was used to assess the correlation of the PI3K/Akt signaling level and catch-up growth with the insulin resistance index (IRI). The values of fasting plasma glucose, fasting insulin and IRI were significantly higher, whereas insulin sensitivity index was significantly lower in IUGR offsprings than those in the controls. The PI3K mRNA and protein levels as well as the phospho-AktSer473 levels were significantly lower in IUGR offsprings compared to the controls. Reductions of GLUT4 as well as increases of PTEN and nuclear fractional PPARγ were detected in IUGR offsprings. Catch-up growth IUGR rats were positively correlated with insulin resistance and underwent more remarkable alterations of the PI3K, PTEN and GLUT4 expressions. Our results demonstrated that rats born IUGR developed insulin resistance later in life, which was likely mediated by reductions of the PI3K/Akt related signaling activities, particularly in those with excess catch-up growth.
Collapse
Affiliation(s)
- Yan Xing
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Jin Zhang
- Department of pediatrics, Beijing Jishuitan Hospital, Beijing, China
| | - Hongling Wei
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Hui Zhang
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| | - Yuhong Guan
- Department of Pulmonary, Beijing Children’s Hospital, Capital Medical University, Beijing, China
| | - Xinli Wang
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
- * E-mail: (XW); (XT)
| | - Xiaomei Tong
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
- * E-mail: (XW); (XT)
| |
Collapse
|
38
|
Altuve M, Severeyn E. Joint analysis of fasting and postprandial plasma glucose and insulin concentrations in Venezuelan women. Diabetes Metab Syndr 2019; 13:2242-2248. [PMID: 31235164 DOI: 10.1016/j.dsx.2019.05.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 05/24/2019] [Indexed: 01/21/2023]
Abstract
AIMS Plasma glucose and insulin concentrations in fasting and postprandial reflect the metabolism of glucose by the human body and are useful in the diagnosis of metabolic diseases, such as diabetes and insulin resistance. In this work, these concentrations are jointly analyzed in Venezuelan women and 28 classes that better specify each metabolic condition are generated. MATERIALS AND METHODS Each class comprises a combination of fasting and postprandial ranges of glucose and insulin concentrations defined in the literature as normal, impaired and diabetic. A hypothesis test was used to find statistically significant differences between the classes, and confidence intervals for age and glucose and insulin concentrations were defined for each class. RESULTS AND CONCLUSION The process of deterioration of glucose metabolism advances with the age of the subject, more than half of the prediabetics have impaired glucose levels in fasting but normal in postprandial and normal insulin levels in fasting and postprandial, and one third of diabetics have diabetic glucose levels in fasting and postprandial and normal insulin levels in fasting and postprandial. This categorization of subjects would allow the application of a more specific treatment and the possibility of predicting the progress of the metabolic disorder.
Collapse
Affiliation(s)
- Miguel Altuve
- Faculty of Electrical and Electronic Engineering, Pontifical Bolivarian University, Bucaramanga, Colombia.
| | - Erika Severeyn
- Department of Thermodynamics and Transfer Phenomena, Simon Bolivar University, Caracas, Venezuela.
| |
Collapse
|
39
|
Hancock ML, Meyer RC, Mistry M, Khetani RS, Wagschal A, Shin T, Ho Sui SJ, Näär AM, Flanagan JG. Insulin Receptor Associates with Promoters Genome-wide and Regulates Gene Expression. Cell 2019; 177:722-736.e22. [PMID: 30955890 PMCID: PMC6478446 DOI: 10.1016/j.cell.2019.02.030] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/07/2019] [Accepted: 02/19/2019] [Indexed: 02/08/2023]
Abstract
Insulin receptor (IR) signaling is central to normal metabolic control and dysregulated in prevalent chronic diseases. IR binds insulin at the cell surface and transduces rapid signaling via cytoplasmic kinases. However, mechanisms mediating long-term effects of insulin remain unclear. Here, we show that IR associates with RNA polymerase II in the nucleus, with striking enrichment at promoters genome-wide. The target genes were highly enriched for insulin-related functions including lipid metabolism and protein synthesis and diseases including diabetes, neurodegeneration, and cancer. IR chromatin binding was increased by insulin and impaired in an insulin-resistant disease model. Promoter binding by IR was mediated by coregulator host cell factor-1 (HCF-1) and transcription factors, revealing an HCF-1-dependent pathway for gene regulation by insulin. These results show that IR interacts with transcriptional machinery at promoters and identify a pathway regulating genes linked to insulin's effects in physiology and disease.
Collapse
Affiliation(s)
- Melissa L. Hancock
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Present address: John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge,
MA, USA
| | - Rebecca C. Meyer
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,These authors contributed equally
| | - Meeta Mistry
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA,These authors contributed equally
| | - Radhika S. Khetani
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA,These authors contributed equally
| | - Alexandre Wagschal
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Present address: Exonics Therapeutics, Cambridge, MA, USA
| | - Taehwan Shin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Shannan J. Ho Sui
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Anders M. Näär
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA,Present address: Department of Nutritional Sciences & Toxicology, University of California, Berkeley, CA
94720, USA
| | - John G. Flanagan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA,Program in Neuroscience, Harvard Medical School, Boston, MA 02115, USA,Lead Contact,Correspondence:
| |
Collapse
|
40
|
Xia H, Dufour CR, Giguère V. ERRα as a Bridge Between Transcription and Function: Role in Liver Metabolism and Disease. Front Endocrinol (Lausanne) 2019; 10:206. [PMID: 31024446 PMCID: PMC6459935 DOI: 10.3389/fendo.2019.00206] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/13/2019] [Indexed: 01/01/2023] Open
Abstract
As transcriptional factors, nuclear receptors (NRs) function as major regulators of gene expression. In particular, dysregulation of NR activity has been shown to significantly alter metabolic homeostasis in various contexts leading to metabolic disorders and cancers. The orphan estrogen-related receptor (ERR) subfamily of NRs, comprised of ERRα, ERRβ, and ERRγ, for which a natural ligand has yet to be identified, are known as central regulators of energy metabolism. If AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin (mTOR) can be viewed as sensors of the metabolic needs of a cell and responding acutely via post-translational control of proteins, then the ERRs can be regarded as downstream effectors of metabolism via transcriptional regulation of genes for a long-term and sustained adaptive response. In this review, we will focus on recent findings centered on the transcriptional roles played by ERRα in hepatocytes. Modulation of ERRα activity in both in vitro and in vivo models via genetic or pharmacological manipulation coupled with chromatin-immunoprecipitation (ChIP)-on-chip and ChIP-sequencing (ChIP-seq) studies have been fundamental in delineating the direct roles of ERRα in the control of hepatic gene expression. These studies have identified crucial roles for ERRα in lipid and carbohydrate metabolism as well as in mitochondrial function under both physiological and pathological conditions. The regulation of ERRα expression and activity via ligand-independent modes of action including coregulator binding, post-translational modifications (PTMs) and control of protein stability will be discussed in the context that may serve as valuable tools to modulate ERRα function as new therapeutic avenues for the treatment of hepatic metabolic dysfunction and related diseases.
Collapse
Affiliation(s)
- Hui Xia
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | | | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, Montréal, QC, Canada
- Department of Biochemistry, McGill University, Montréal, QC, Canada
- Medicine and Oncology, McGill University, Montréal, QC, Canada
| |
Collapse
|
41
|
Wang Z, Dong C. Gluconeogenesis in Cancer: Function and Regulation of PEPCK, FBPase, and G6Pase. Trends Cancer 2018; 5:30-45. [PMID: 30616754 DOI: 10.1016/j.trecan.2018.11.003] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 01/19/2023]
Abstract
Cancer cells display a high rate of glycolysis in the presence of oxygen to promote proliferation. Gluconeogenesis, the reverse pathway of glycolysis, can antagonize aerobic glycolysis in cancer via three key enzymes - phosphoenolpyruvate carboxykinase (PEPCK), fructose-1,6-bisphosphatase (FBPase), and glucose-6-phosphatase (G6Pase). Recent studies have revealed that, in addition to metabolic regulation, these enzymes also play a role in signaling, proliferation, and the cancer stem cell (CSC) tumor phenotype. Multifaceted regulation of PEPCK, FBPase, and G6Pase through transcription, epigenetics, post-translational modification, and enzymatic activity is observed in different cancers. We review here the function and regulation of key gluconeogenic enzymes and new therapeutic opportunities.
Collapse
Affiliation(s)
- Zhanyu Wang
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (Breast Center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, and Department of Surgical Oncology (Breast Center) of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
42
|
Rajaie A, Allahyari M, Nazari-Robati M, Fallah H. Inhibition of Interleukin-1 Receptor-Associated Kinases 1/4, Increases Gene Expression and Serum Level of Adiponectin in Mouse Model of Insulin Resistance. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2018; 7:185-192. [PMID: 31565650 PMCID: PMC6744615 DOI: 10.22088/ijmcm.bums.7.3.185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 10/08/2018] [Indexed: 11/07/2022]
Abstract
Insulin resistance is a feature of most patients with type 2 diabetes mellitus. Epidemiological evidence suggest a correlation between inflammation and insulin resistant states such as obesity, but the underlying mechanisms are largely unknown. Interleukin-1 receptor-associated kinases (IRAK) play a central role in inflammatory responses by regulating the expression of various inflammatory genes in immune cells. This study was aimed to investigate the effect of IRAK inhibitor on gene transcription and serum concentration of adiponectin in insulin-resistant mice. Experimental mice were randomly divided into 6 groups: the healthy control group was fed a regular chow diet while other groups were fed with a high-fat diet for 12 weeks. After this period, the animals were treated with IRAK inhibitor, pioglitazone, both IRAK and pioglitazone, and DMSO, for two weeks. Adiponectin gene expression level was analyzed by real-time PCR. Additionally, serum adiponectin levels were measured by ELISA. Homeostasis model assessment-adiponectin (HOMA-AD) as an insulin sensitivity index was calculated. IRAK inhibitor and pioglitazone increased significantly the expression of adiponectin gene. Also, adiponectin concentration in the control group (9.67±1.1 μg/ml) increased to 25.34±2.04 μg/ml in pioglitazone treatment group. IRAK inhibitor also increased adiponectin concentration (18.24±1.53 μg/ml) but did not show a synergistic effect with pioglitazone when administered simultaneously (26.66±2.5 μg/ml). HOMA-AD was 0.33±0.04 in pioglitazone treated group, 0.6±0.13 in IRAK inhibitor group, and 0.31±0.03 in animals that received IRAKi and pioglitazone. Our findings suggest that increased adiponectin secretion from adipose tissue mediated by IRAK inhibitor may increase the insulin sensitivity in an animal model of insulin resistance.
Collapse
Affiliation(s)
- Athena Rajaie
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mostafa Allahyari
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdieh Nazari-Robati
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Hossein Fallah
- Department of Clinical Biochemistry, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
43
|
Yan J, Wang C, Jin Y, Meng Q, Liu Q, Liu Z, Liu K, Sun H. Catalpol ameliorates hepatic insulin resistance in type 2 diabetes through acting on AMPK/NOX4/PI3K/AKT pathway. Pharmacol Res 2018; 130:466-480. [DOI: 10.1016/j.phrs.2017.12.026] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 12/11/2017] [Accepted: 12/22/2017] [Indexed: 12/23/2022]
|
44
|
You D, Nilsson E, Tenen DE, Lyubetskaya A, Lo JC, Jiang R, Deng J, Dawes BA, Vaag A, Ling C, Rosen ED, Kang S. Dnmt3a is an epigenetic mediator of adipose insulin resistance. eLife 2017; 6:30766. [PMID: 29091029 PMCID: PMC5730374 DOI: 10.7554/elife.30766] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 10/29/2017] [Indexed: 12/21/2022] Open
Abstract
Insulin resistance results from an intricate interaction between genetic make-up and environment, and thus may be orchestrated by epigenetic mechanisms like DNA methylation. Here, we demonstrate that DNA methyltransferase 3a (Dnmt3a) is both necessary and sufficient to mediate insulin resistance in cultured mouse and human adipocytes. Furthermore, adipose-specific Dnmt3a knock-out mice are protected from diet-induced insulin resistance and glucose intolerance without accompanying changes in adiposity. Unbiased gene profiling studies revealed Fgf21 as a key negatively regulated Dnmt3a target gene in adipocytes with concordant changes in DNA methylation at the Fgf21 promoter region. Consistent with this, Fgf21 can rescue Dnmt3a-mediated insulin resistance, and DNA methylation at the FGF21 locus was elevated in human subjects with diabetes and correlated negatively with expression of FGF21 in human adipose tissue. Taken together, our data demonstrate that adipose Dnmt3a is a novel epigenetic mediator of insulin resistance in vitro and in vivo.
Collapse
Affiliation(s)
- Dongjoo You
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| | - Emma Nilsson
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Danielle E Tenen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, United States
| | | | - James C Lo
- Weill Cornell Medical College, New York, United States
| | - Rencong Jiang
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| | - Jasmine Deng
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| | - Brian A Dawes
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, United States
| | - Allan Vaag
- Diabetes and Metabolism, Department of Endocrinology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,Early Clinical Development, AstraZeneca, Innovative Medicines, Göteborg, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes Unit, Department of Clinical Sciences, Lund University Diabetes Centre, Scania University Hospital, Malmö, Sweden
| | - Evan D Rosen
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Boston, United States.,Broad Institute of Harvard and MIT, Cambridge, United States
| | - Sona Kang
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
45
|
Lark DS, Wasserman DH. Meta-fibrosis links positive energy balance and mitochondrial metabolism to insulin resistance. F1000Res 2017; 6:1758. [PMID: 29043068 PMCID: PMC5621108 DOI: 10.12688/f1000research.11653.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/28/2017] [Indexed: 12/12/2022] Open
Abstract
Obesity and insulin resistance often emerge from positive energy balance and generally are linked to low-grade inflammation. This low-grade inflammation has been called “meta-inflammation” because it is a consequence of the metabolic dysregulation that can accompany overnutrition. One means by which meta-inflammation is linked to insulin resistance is extracellular matrix expansion secondary to meta-inflammation, which we define here as “meta-fibrosis”. The significance of meta-fibrosis is that it reflects a situation in which the extracellular matrix functions as a multi-level integrator of local (for example, mitochondrial reactive oxygen species production) and systemic (for example, inflammation) inputs that couple to cellular processes creating insulin resistance. While adipose tissue extracellular matrix remodeling has received considerable attention, it is becoming increasingly apparent that liver and skeletal muscle extracellular matrix remodeling also contributes to insulin resistance. In this review, we address recent advances in our understanding of energy balance, mitochondrial energetics, meta-inflammation, and meta-fibrosis in the development of insulin resistance.
Collapse
Affiliation(s)
- Daniel S Lark
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology & Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA.,Vanderbilt Mouse Metabolic Phenotyping Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
46
|
Lee JY, Tae JC, Kim CH, Hwang D, Kim KC, Suh CS, Kim SH. Expression of the genes for peroxisome proliferator-activated receptor-γ, cyclooxygenase-2, and proinflammatory cytokines in granulosa cells from women with polycystic ovary syndrome. Clin Exp Reprod Med 2017; 44:146-151. [PMID: 29026721 PMCID: PMC5636927 DOI: 10.5653/cerm.2017.44.3.146] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/20/2017] [Accepted: 06/19/2017] [Indexed: 12/14/2022] Open
Abstract
Objective To identify differences in the expression of the genes for peroxisome proliferator-activated receptor (PPAR)-γ, cyclooxygenase (COX)-2, and the proinflammatory cytokines interleukin (IL)-6 and tumor necrosis factor (TNF)-α in granulosa cells (GCs) from polycystic ovary syndrome (PCOS) patients and controls undergoing controlled ovarian stimulation. Methods Nine patients with PCOS and six controls were enrolled in this study. On the day of oocyte retrieval, GCs were collected from pooled follicular fluid. Total mRNA was extracted from GCs. Reverse transcription was performed and gene expression levels were quantified by realtime quantitative polymerase chain reaction. Results There were no significant differences in age, body mass index, and total gonadotropin dose, except for the ratio of luteinizing hormone to follicle-stimulating hormone between the PCOS and control groups. PPAR-γ and COX-2 mRNA was significantly downregulated in the GCs of PCOS women compared with controls (p=0.034 and p=0.018, respectively), but the expression of IL-6 and TNF-α mRNA did not show significant differences. No significant correlation was detected between the expression of these mRNA sequences and clinical characteristics, including the number of retrieved oocytes, oocyte maturity, cleavage, or the good embryo rate. Positive correlations were found among the PPAR-γ, COX-2, IL-6, and TNF-α mRNA levels. Conclusion Our data may provide novel clues regarding ovarian GC dysfunction in PCOS, and indirectly provide evidence that the effect of PPAR-γ agonists in PCOS might result from alterations in the ovarian follicular environment. Further studies with a larger sample size are required to confirm these proposals.
Collapse
Affiliation(s)
| | | | | | | | | | - Chang Suk Suh
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| | - Seok Hyun Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
47
|
Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017; 23:804-814. [PMID: 28697184 DOI: 10.1038/nm.4350] [Citation(s) in RCA: 782] [Impact Index Per Article: 111.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 05/11/2017] [Indexed: 12/12/2022]
Abstract
Nutritional excess is a major forerunner of type 2 diabetes. It enhances the secretion of insulin, but attenuates insulin's metabolic actions in the liver, skeletal muscle and adipose tissue. However, conflicting evidence indicates a lack of knowledge of the timing of these events during the development of obesity and diabetes, pointing to a key gap in our understanding of metabolic disease. This Perspective reviews alternate viewpoints and recent results on the temporal and mechanistic connections between hyperinsulinemia, obesity and insulin resistance. Although much attention has addressed early steps in the insulin signaling cascade, insulin resistance in obesity seems to be largely elicited downstream of these steps. New findings also connect insulin resistance to extensive metabolic cross-talk between the liver, adipose tissue, pancreas and skeletal muscle. These and other advances over the past 5 years offer exciting opportunities and daunting challenges for the development of new therapeutic strategies for the treatment of type 2 diabetes.
Collapse
|
48
|
Pioglitazone and cardiovascular risk reduction: time for a second look? Cardiovasc Endocrinol 2017; 6:55-61. [PMID: 31646121 DOI: 10.1097/xce.0000000000000110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/17/2016] [Indexed: 11/26/2022] Open
Abstract
Insulin resistance, a fundamental pathophysiological abnormality in patients with type 2 diabetes, is associated with increased cardiovascular (CV) disease risk. In diabetes management, the macrovascular impact of antihyperglycemic agents that do not improve insulin sensitivity has generally been disappointing. In contrast, glucose-lowering drugs that work as insulin sensitizing agents have been postulated to reduce CV complications. The data to support this hypothesis have, however, been inconsistent. The impact of thiazolidinediones on macrovascular events is of particular interest. In this review, we discuss the results of trials reporting CV outcomes in patients treated with thiazolidinediones. We focus on the findings of the recent Insulin Resistance Intervention after Stroke trial that demonstrated a beneficial effect of pioglitazone on CV outcomes in stroke patients with insulin resistance. We discuss the Insulin Resistance Intervention after Stroke results and its implications for clinical practice. We discuss the selective use of pioglitazone as secondary prevention to reduce CV risk in insulin resistant patients.
Collapse
|
49
|
Li H, Qin T, Li M, Ma S. Thymol improves high-fat diet-induced cognitive deficits in mice via ameliorating brain insulin resistance and upregulating NRF2/HO-1 pathway. Metab Brain Dis 2017; 32:385-393. [PMID: 27761760 DOI: 10.1007/s11011-016-9921-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 10/06/2016] [Indexed: 01/08/2023]
Abstract
The impaired insulin signaling has been recognized as a common pathogenetic mechanism between diabetes and Alzheimer's disease (AD). In the progression of AD, brain is characterized by defective insulin receptor substrate-1 (IRS-1) and increased oxidative stress. Thymol, a monoterpene phenol isolated from medicinal herbs, has exhibited robust neuroprotective effects. The present study was designed to investigate the protective effect of thymol on HFD-induced cognitive deficits, and explore the possible mechanisms. C57BL/6 J mice were fed for 12 weeks with either HFD or normal diet. The mice fed with HFD were dosed with metformin (200 mg/kg) or thymol (20, 40 mg/kg) daily. It was observed that thymol treatment significantly reversed the gain of body weight and peripheral insulin resistance induced by HFD. Meanwhile, thymol improved the cognitive impairments in the Morris Water Maze (MWM) test and decreased HFD-induced Aβ deposition and tau hyperphosphorylation in the hippocampus, which may be correlated with the inhibition of hippocampal oxidative stress and inflammation. In addition, thymol down-regulated the level of P-Ser307 IRS-1, and hence enhancing the expression of P-Ser473 AKT and P-Ser9 GSK3β. We further found that the protective effects of thymol on cognitive impairments were associated with the up-regulation of nuclear respiratory factor (Nrf2)/heme oxygenase-1(HO-1) pathway. In conclusion, thymol exhibited beneficial effects on HFD-induced cognitive deficits through improving hippocampal insulin resistance, and activating Nrf2/HO-1 signaling.
Collapse
Affiliation(s)
- Hongyan Li
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China
| | - Tingting Qin
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China
| | - Min Li
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China
| | - Shiping Ma
- Department of Pharmacology of Chinese Materia Medica, China PharmaceuticalUniversity, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
50
|
The Na+-D-glucose cotransporters SGLT1 and SGLT2 are targets for the treatment of diabetes and cancer. Pharmacol Ther 2017; 170:148-165. [DOI: 10.1016/j.pharmthera.2016.10.017] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|