1
|
Goldberg D, Buchshtab N, Charni-Natan M, Goldstein I. Transcriptional cascades during fasting amplify gluconeogenesis and instigate a secondary wave of ketogenic gene transcription. Liver Int 2024; 44:2964-2982. [PMID: 39162082 DOI: 10.1111/liv.16077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/04/2024] [Accepted: 08/09/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND AND AIMS During fasting, bodily homeostasis is maintained due to hepatic production of glucose (gluconeogenesis) and ketone bodies (ketogenesis). The main hormones governing hepatic fuel production are glucagon and glucocorticoids that initiate transcriptional programs aimed at supporting gluconeogenesis and ketogenesis. METHODS Using primary mouse hepatocytes as an ex vivo model, we employed transcriptomic analysis (RNA-seq), genome-wide profiling of enhancer dynamics (ChIP-seq), perturbation experiments (inhibitors, shRNA), hepatic glucose production measurements and computational analyses. RESULTS We found that in addition to the known metabolic genes transcriptionally induced by glucagon and glucocorticoids, these hormones induce a set of genes encoding transcription factors (TFs) thereby initiating transcriptional cascades. Upon activation by glucocorticoids, the glucocorticoid receptor (GR) induced the genes encoding two TFs: CCAAT/enhancer-binding protein beta (C/EBPβ) and peroxisome proliferator-activated receptor alpha (PPARα). We found that the GR-C/EBPβ cascade mainly serves as a secondary amplifier of primary hormone-induced gene programs. C/EBPβ augmented gluconeogenic gene expression and hepatic glucose production. Conversely, the GR-PPARα cascade initiated a secondary transcriptional wave of genes supporting ketogenesis. The cascade led to synergistic induction of ketogenic genes which is dependent on protein synthesis. Genome-wide analysis of enhancer dynamics revealed numerous enhancers activated by the GR-PPARα cascade. These enhancers were proximal to ketogenic genes, enriched for the PPARα response element and showed increased PPARα binding. CONCLUSION This study reveals abundant transcriptional cascades occurring during fasting. These cascades serve two separated purposes: the amplification of the gluconeogenic transcriptional program and the induction of a gene program aimed at enhancing ketogenesis.
Collapse
Affiliation(s)
- Dana Goldberg
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Nufar Buchshtab
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Meital Charni-Natan
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Ido Goldstein
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
2
|
Gupta I, Gaykalova DA. Unveiling the role of PIK3R1 in cancer: A comprehensive review of regulatory signaling and therapeutic implications. Semin Cancer Biol 2024; 106-107:58-86. [PMID: 39197810 DOI: 10.1016/j.semcancer.2024.08.004] [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: 05/07/2024] [Revised: 07/11/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Phosphoinositide 3-kinase (PI3K) is responsible for phosphorylating phosphoinositides to generate secondary signaling molecules crucial for regulating various cellular processes, including cell growth, survival, and metabolism. The PI3K is a heterodimeric enzyme complex comprising of a catalytic subunit (p110α, p110β, or p110δ) and a regulatory subunit (p85). The binding of the regulatory subunit, p85, with the catalytic subunit, p110, forms an integral component of the PI3K enzyme. PIK3R1 (phosphoinositide-3-kinase regulatory subunit 1) belongs to class IA of the PI3K family. PIK3R1 exhibits structural complexity due to alternative splicing, giving rise to distinct isoforms, prominently p85α and p55α. While the primary p85α isoform comprises multiple domains, including Src homology 3 (SH3) domains, a Breakpoint Cluster Region Homology (BH) domain, and Src homology 2 (SH2) domains (iSH2 and nSH2), the shorter isoform, p55α, lacks certain domains present in p85α. In this review, we will highlight the intricate regulatory mechanisms governing PI3K signaling along with the impact of PIK3R1 alterations on cellular processes. We will further delve into the clinical significance of PIK3R1 mutations in various cancer types and their implications for prognosis and treatment outcomes. Additionally, we will discuss the evolving landscape of targeted therapies aimed at modulating PI3K-associated pathways. Overall, this review will provide insights into the dynamic interplay of PIK3R1 in cancer, fostering advancements in precision medicine and the development of targeted interventions.
Collapse
Affiliation(s)
- Ishita Gupta
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA
| | - Daria A Gaykalova
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA; Department of Otorhinolaryngology-Head and Neck Surgery, Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD, USA; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
3
|
Zhou F, Sheng C, Ma X, Li T, Ming X, Wang S, Tan J, Yang Y, Sun H, Lu J, Liu J, Deng R, Wang X, Zhou L. BCKDH kinase promotes hepatic gluconeogenesis independent of BCKDHA. Cell Death Dis 2024; 15:736. [PMID: 39389936 PMCID: PMC11467410 DOI: 10.1038/s41419-024-07071-0] [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: 03/30/2024] [Revised: 09/01/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024]
Abstract
Elevated circulating branched-chain amino acids (BCAAs) are tightly linked to an increased risk in the development of type 2 diabetes mellitus. The rate limiting enzyme of BCAA catabolism branched-chain α-ketoacid dehydrogenase (BCKDH) is phosphorylated at E1α subunit (BCKDHA) by its kinase (BCKDK) and inactivated. Here, the liver-specific BCKDK or BCKDHA knockout mice displayed normal glucose tolerance and insulin sensitivity. However, knockout of BCKDK in the liver inhibited hepatic glucose production as well as the expression of key gluconeogenic enzymes. No abnormal gluconeogenesis was found in mice lacking hepatic BCKDHA. Consistent with the vivo results, BT2-mediated inhibition or genetic knockdown of BCKDK decreased hepatic glucose production and gluconeogenic gene expressions in primary mouse hepatocytes while BCKDK overexpression exhibited an opposite effect. Whereas, gluconeogenic gene expressions were not altered in BCKDHA-silenced hepatocytes. Mechanistically, BT2 treatment attenuated the interaction of cAMP response element binding protein (CREB) with CREB-binding protein and promoted FOXO1 protein degradation by increasing its ubiquitination. Our findings suggest that BCKDK regulates hepatic gluconeogenesis through CREB and FOXO1 signalings, independent of BCKDHA-mediated BCAA catabolism.
Collapse
Affiliation(s)
- Feiye Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Department of Endocrine and Metabolic Diseases, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Chunxiang Sheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoqin Ma
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tianjiao Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xing Ming
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shushu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jialin Tan
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yulin Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Haipeng Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Center for Cardiovascular Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, 300134, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianmin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruyuan Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, 200032, China; Shanghai Institute of Liver Disease, Shanghai, 200032, China.
| | - Xiao Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Libin Zhou
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
4
|
Nie W, Zhu H, Sun X, Zhou J, Xu H, Yu Z, Lu M, Jiang B, Zhou L, Zhou X. Catalpol attenuates hepatic glucose metabolism disorder and oxidative stress in triptolide-induced liver injury by regulating the SIRT1/HIF-1α pathway. Int J Biol Sci 2024; 20:4077-4097. [PMID: 39113710 PMCID: PMC11302874 DOI: 10.7150/ijbs.97362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
Triptolide (TP), known for its effectiveness in treating various rheumatoid diseases, is also associated with significant hepatotoxicity risks. This study explored Catalpol (CAT), an iridoid glycoside with antioxidative and anti-inflammatory effects, as a potential defense against TP-induced liver damage. In vivo and in vitro models of liver injury were established using TP in combination with different concentrations of CAT. Metabolomics analyses were conducted to assess energy metabolism in mouse livers. Additionally, a Seahorse XF Analyzer was employed to measure glycolysis rate, mitochondrial respiratory functionality, and real-time ATP generation rate in AML12 cells. The study also examined the expression of proteins related to glycogenolysis and gluconeogenesis. Using both in vitro SIRT1 knockout/overexpression and in vivo liver-specific SIRT1 knockout models, we confirmed SIRT1 as a mechanism of action for CAT. Our findings revealed that CAT could alleviate TP-induced liver injury by activating SIRT1, which inhibited lysine acetylation of hypoxia-inducible factor-1α (HIF-1α), thereby restoring the balance between glycolysis and oxidative phosphorylation. This action improved mitochondrial dysfunction and reduced glucose metabolism disorder and oxidative stress caused by TP. Taken together, these insights unveil a hitherto undocumented mechanism by which CAT ameliorates TP-induced liver injury, positioning it as a potential therapeutic agent for managing TP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Weijue Nie
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hong Zhu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xin Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jie Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Heng Xu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zhichao Yu
- School of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Minghao Lu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Baoping Jiang
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Lingling Zhou
- Department of Pharmacology, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xueping Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
5
|
Kajani S, Laker RC, Ratkova E, Will S, Rhodes CJ. Hepatic glucagon action: beyond glucose mobilization. Physiol Rev 2024; 104:1021-1060. [PMID: 38300523 DOI: 10.1152/physrev.00028.2023] [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: 07/11/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 02/02/2024] Open
Abstract
Glucagon's ability to promote hepatic glucose production has been known for over a century, with initial observations touting this hormone as a diabetogenic agent. However, glucagon receptor agonism [when balanced with an incretin, including glucagon-like peptide 1 (GLP-1) to dampen glucose excursions] is now being developed as a promising therapeutic target in the treatment of metabolic diseases, like metabolic dysfunction-associated steatotic disease/metabolic dysfunction-associated steatohepatitis (MASLD/MASH), and may also have benefit for obesity and chronic kidney disease. Conventionally regarded as the opposing tag-team partner of the anabolic mediator insulin, glucagon is gradually emerging as more than just a "catabolic hormone." Glucagon action on glucose homeostasis within the liver has been well characterized. However, growing evidence, in part thanks to new and sensitive "omics" technologies, has implicated glucagon as more than just a "glucose liberator." Elucidation of glucagon's capacity to increase fatty acid oxidation while attenuating endogenous lipid synthesis speaks to the dichotomous nature of the hormone. Furthermore, glucagon action is not limited to just glucose homeostasis and lipid metabolism, as traditionally reported. Glucagon plays key regulatory roles in hepatic amino acid and ketone body metabolism, as well as mitochondrial turnover and function, indicating broader glucagon signaling consequences for metabolic homeostasis mediated by the liver. Here we examine the broadening role of glucagon signaling within the hepatocyte and question the current dogma, to appreciate glucagon as more than just that "catabolic hormone."
Collapse
Affiliation(s)
- Sarina Kajani
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Rhianna C Laker
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Ekaterina Ratkova
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Mölndal, Sweden
| | - Sarah Will
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| | - Christopher J Rhodes
- Early Cardiovascular, Renal and Metabolism, Biopharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, United States
| |
Collapse
|
6
|
Cao X, Chen L, Lu K, Yu T, Xia H, Wang S, Sun G, Liu P, Liao W. Egg white-derived peptides reduced blood glucose in high-fat-diet and low-dose streptozotocin-induced type 2 diabetic mice via regulating the hepatic gluconeogenic signaling and metabolic profile. Food Funct 2024; 15:7003-7016. [PMID: 38855929 DOI: 10.1039/d4fo00725e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Food proteins are considered an ideal source for the identification of bioactive peptides with the potential to intervene in nutrition-related chronic diseases such as cardiovascular disease, obesity, and diabetes. Egg white-derived peptides (EWPs) have been shown to improve glucose tolerance in insulin-resistant rats. However, underlying mechanisms are to be elucidated. Therefore, we hypothesized that EWP exerts a hypoglycemic effect by regulating hepatic glucose homeostasis. Our results showed that 7 weeks of EWP treatment reduced the fasting blood glucose in T2DM mice and the inhibition of the liver gluconeogenic pathway was involved in the mechanisms of actions. Using the untargeted metabolomics technique, we found that EWP treatment also altered the hepatic metabolic profile in T2DM mice, in which, the role of fatty acid esters of hydroxy fatty acids in mediating the hypoglycemic effect of EWPs might be pivotal.
Collapse
Affiliation(s)
- Xinyi Cao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, P.R. China.
| | - Liang Chen
- Public Service Platform of South China Sea for R&D Marine Biomedicine Resources, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, 524023, China
| | - Kun Lu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, P.R. China.
| | - Tingqing Yu
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, P.R. China.
| | - Hui Xia
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, P.R. China.
| | - Shaokang Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, P.R. China.
| | - Guiju Sun
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, P.R. China.
| | - Ping Liu
- Department of Food Science and Technology, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou, 225300, P.R. China
| | - Wang Liao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing 210009, P.R. China.
| |
Collapse
|
7
|
Ou K, Zhang Q, Xi F, Ni H, Lu J, Lyu X, Wang C, Li Q, Wang Q. Prenatal EGCG consumption impacts hepatic glycogen synthesis and lipid metabolism in adult mice. Int J Biol Macromol 2024; 260:129491. [PMID: 38228202 DOI: 10.1016/j.ijbiomac.2024.129491] [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: 10/16/2023] [Revised: 12/27/2023] [Accepted: 01/12/2024] [Indexed: 01/18/2024]
Abstract
In this study, the impact of prenatal exposure to Epigallocatechin gallate (EGCG) on the liver of adult offspring mice was investigated. While EGCG is known for its health benefits, its effects of prenatal exposure on the liver remain unclear. Pregnant C57BL/6 J mice were exposed to 1 mg/kg of EGCG for 16 days to assess hepatotoxicity effects of adult offspring. Transcriptomics and metabolomics were employed to elucidate the hepatotoxicity mechanisms. The findings revealed that prenatal EGCG exposure led to a decrease in liver somatic index, enhanced inflammatory responses and disrupted liver function through increased glycogen accumulation in adult mice. The integrated omics analysis revealed significant alterations in key pathways involved in liver glucose lipid metabolism, such as gluconeogenesis, dysregulation of insulin signaling, and induction of liver inflammation. Furthermore, the study found a negative correlation between the promoter methylation levels of Ppara and their mRNA levels, suggesting that EGCG could reduce hepatic lipid content through epigenetic modifications. The findings suggest that prenatal EGCG exposure can have detrimental impacts on the liver among adult individuals and emphasize the need for a comprehensive evaluation of the potential risks associated with EGCG consumption during pregnancy.
Collapse
Affiliation(s)
- Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Quan Zhang
- School of Medicine, Xiamen University, Xiamen, Fujian 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Feifei Xi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Huizhen Ni
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Jiebo Lu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Xuejing Lyu
- School of Medicine, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Qiyuan Li
- School of Medicine, Xiamen University, Xiamen, Fujian 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian 361102, China; Department of Pediatrics, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian 361003, China.
| | - Qin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361005, PR China.
| |
Collapse
|
8
|
Pan Q, Ai W, Guo S. TGF-β1 Signaling Impairs Metformin Action on Glycemic Control. Int J Mol Sci 2024; 25:2424. [PMID: 38397103 PMCID: PMC10889280 DOI: 10.3390/ijms25042424] [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: 01/07/2024] [Revised: 02/02/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024] Open
Abstract
Hyperglycemia is a hallmark of type 2 diabetes (T2D). Metformin, the first-line drug used to treat T2D, maintains blood glucose within a normal range by suppressing hepatic glucose production (HGP). However, resistance to metformin treatment is developed in most T2D patients over time. Transforming growth factor beta 1 (TGF-β1) levels are elevated both in the liver and serum of T2D humans and mice. Here, we found that TGF-β1 treatment impairs metformin action on suppressing HGP via inhibiting AMPK phosphorylation at Threonine 172 (T172). Hepatic TGF-β1 deficiency improves metformin action on glycemic control in high fat diet (HFD)-induced obese mice. In our hepatic insulin resistant mouse model (hepatic insulin receptor substrate 1 (IRS1) and IRS2 double knockout (DKO)), metformin action on glycemic control was impaired, which is largely improved by further deletion of hepatic TGF-β1 (TKObeta1) or hepatic Foxo1 (TKOfoxo1). Moreover, blockade of TGF-β1 signaling by chemical inhibitor of TGF-β1 type I receptor LY2157299 improves to metformin sensitivity in mice. Taken together, our current study suggests that hepatic TGF-β1 signaling impairs metformin action on glycemic control, and suppression of TGF-β1 signaling could serve as part of combination therapy with metformin for T2D treatment.
Collapse
Affiliation(s)
| | | | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA; (Q.P.); (W.A.)
| |
Collapse
|
9
|
Zhou L, Su W, Wang Y, Zhang Y, Xia Z, Lei S. FOXO1 reduces STAT3 activation and causes impaired mitochondrial quality control in diabetic cardiomyopathy. Diabetes Obes Metab 2024; 26:732-744. [PMID: 37961034 DOI: 10.1111/dom.15369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 10/30/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
AIMS To investigate the role of FOXO1 in STAT3 activation and mitochondrial quality control in the diabetic heart. METHODS Type 1 diabetes mellitus (T1DM) was induced in rats by a single intraperitoneal injection of 60 mg · kg-1 streptozotocin (STZ), while type 2 diabetes mellitus (T2DM) was induced in rats with a high-fat diet through intraperitoneal injection of 35 mg · kg-1 STZ. Primary neonatal mouse cardiomyocytes and H9c2 cells were exposed to low glucose (5.5 mM) or high glucose (HG; 30 mM) with or without treatment with the FOXO1 inhibitor AS1842856 (1 μM) for 24 hours. In addition, the diabetic db/db mice (aged 8 weeks) and sex- and age-matched non-diabetic db/+ mice were treated with vehicle or AS1842856 by oral gavage for 15 days at a dose of 5 mg · kg-1 · d-1 . RESULTS Rats with T1DM or T2DM had excessive cardiac FOXO1 activation, accompanied by decreased STAT3 activation. Immunofluorescence and immunoprecipitation analysis showed colocalization and association of FOXO1 and STAT3 under basal conditions in isolated cardiomyocytes. Selective inhibition of FOXO1 activation by AS1842856 or FOXO1 siRNA transfection improved STAT3 activation, mitophagy and mitochondrial fusion, and decreased mitochondrial fission in isolated cardiomyocytes exposed to HG. Transfection with STAT3 siRNA further reduced mitophagy, mitochondrial fusion and increased mitochondrial fission in HG-treated cardiomyocytes. AS1842856 alleviated cardiac dysfunction, pathological damage and improved STAT3 activation, mitophagy and mitochondrial dynamics in diabetic db/db mice. Additionally, AS1842856 improved mitochondrial function indicated by increased mitochondrial membrane potential and adenosine triphosphate production and decreased mitochondrial reactive oxygen species production in isolated cardiomyocytes exposed to HG. CONCLUSIONS Excessive FOXO1 activation during diabetes reduces STAT3 activation, with subsequent impairment of mitochondrial quality, ultimately promoting the development of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wating Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yafeng Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuefu Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
10
|
Pfaller AM, Kaplan L, Carido M, Grassmann F, Díaz-Lezama N, Ghaseminejad F, Wunderlich KA, Glänzer S, Bludau O, Pannicke T, Weber BHF, Koch SF, Bonev B, Hauck SM, Grosche A. The glucocorticoid receptor as a master regulator of the Müller cell response to diabetic conditions in mice. J Neuroinflammation 2024; 21:33. [PMID: 38273366 PMCID: PMC10809506 DOI: 10.1186/s12974-024-03021-x] [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: 09/12/2023] [Accepted: 01/11/2024] [Indexed: 01/27/2024] Open
Abstract
Diabetic retinopathy (DR) is considered a primarily microvascular complication of diabetes. Müller glia cells are at the centre of the retinal neurovascular unit and play a critical role in DR. We therefore investigated Müller cell-specific signalling pathways that are altered in DR to identify novel targets for gene therapy. Using a multi-omics approach on purified Müller cells from diabetic db/db mice, we found the mRNA and protein expression of the glucocorticoid receptor (GR) to be significantly decreased, while its target gene cluster was down-regulated. Further, oPOSSUM TF analysis and ATAC- sequencing identified the GR as a master regulator of Müller cell response to diabetic conditions. Cortisol not only increased GR phosphorylation. It also induced changes in the expression of known GR target genes in retinal explants. Finally, retinal functionality was improved by AAV-mediated overexpression of GR in Müller cells. Our study demonstrates an important role of the glial GR in DR and implies that therapeutic approaches targeting this signalling pathway should be aimed at increasing GR expression rather than the addition of more ligand.
Collapse
Affiliation(s)
- Anna M Pfaller
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Lew Kaplan
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Madalena Carido
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Felix Grassmann
- Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
- Institute for Clinical Research and Systems Medicine, Health and Medical University, Potsdam, Germany
| | - Nundehui Díaz-Lezama
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Farhad Ghaseminejad
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Kirsten A Wunderlich
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Institute for Molecular Medicine, Health and Medical University, Potsdam, Germany
| | - Sarah Glänzer
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Oliver Bludau
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Thomas Pannicke
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Bernhard H F Weber
- Institute of Clinical Human Genetics, University Hospital Regensburg, Regensburg, Germany
- Institute of Human Genetics, University Regensburg, Regensburg, Germany
| | - Susanne F Koch
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Boyan Bonev
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Antje Grosche
- Department of Physiological Genomics, Biomedical Center-BMC, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany.
| |
Collapse
|
11
|
Yang W, Jiang W, Guo S. Regulation of Macronutrients in Insulin Resistance and Glucose Homeostasis during Type 2 Diabetes Mellitus. Nutrients 2023; 15:4671. [PMID: 37960324 PMCID: PMC10647592 DOI: 10.3390/nu15214671] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Insulin resistance is an important feature of metabolic syndrome and a precursor of type 2 diabetes mellitus (T2DM). Overnutrition-induced obesity is a major risk factor for the development of insulin resistance and T2DM. The intake of macronutrients plays a key role in maintaining energy balance. The components of macronutrients distinctly regulate insulin sensitivity and glucose homeostasis. Precisely adjusting the beneficial food compound intake is important for the prevention of insulin resistance and T2DM. Here, we reviewed the effects of different components of macronutrients on insulin sensitivity and their underlying mechanisms, including fructose, dietary fiber, saturated and unsaturated fatty acids, and amino acids. Understanding the diet-gene interaction will help us to better uncover the molecular mechanisms of T2DM and promote the application of precision nutrition in practice by integrating multi-omics analysis.
Collapse
Affiliation(s)
| | | | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX 77843, USA; (W.Y.); (W.J.)
| |
Collapse
|
12
|
Teaney NA, Cyr NE. FoxO1 as a tissue-specific therapeutic target for type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1286838. [PMID: 37941908 PMCID: PMC10629996 DOI: 10.3389/fendo.2023.1286838] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/06/2023] [Indexed: 11/10/2023] Open
Abstract
Forkhead box O (FoxO) proteins are transcription factors that mediate many aspects of physiology and thus have been targeted as therapeutics for several diseases including metabolic disorders such as type 2 diabetes mellitus (T2D). The role of FoxO1 in metabolism has been well studied, but recently FoxO1's potential for diabetes prevention and therapy has been debated. For example, studies have shown that increased FoxO1 activity in certain tissue types contributes to T2D pathology, symptoms, and comorbidities, yet in other tissue types elevated FoxO1 has been reported to alleviate symptoms associated with diabetes. Furthermore, studies have reported opposite effects of active FoxO1 in the same tissue type. For example, in the liver, FoxO1 contributes to T2D by increasing hepatic glucose production. However, FoxO1 has been shown to either increase or decrease hepatic lipogenesis as well as adipogenesis in white adipose tissue. In skeletal muscle, FoxO1 reduces glucose uptake and oxidation, promotes lipid uptake and oxidation, and increases muscle atrophy. While many studies show that FoxO1 lowers pancreatic insulin production and secretion, others show the opposite, especially in response to oxidative stress and inflammation. Elevated FoxO1 in the hypothalamus increases the risk of developing T2D. However, increased FoxO1 may mitigate Alzheimer's disease, a neurodegenerative disease strongly associated with T2D. Conversely, accumulating evidence implicates increased FoxO1 with Parkinson's disease pathogenesis. Here we review FoxO1's actions in T2D conditions in metabolic tissues that abundantly express FoxO1 and highlight some of the current studies targeting FoxO1 for T2D treatment.
Collapse
Affiliation(s)
- Nicole A. Teaney
- Stonehill College, Neuroscience Program, Easton, MA, United States
| | - Nicole E. Cyr
- Stonehill College, Neuroscience Program, Easton, MA, United States
- Stonehill College, Department of Biology, Easton, MA, United States
| |
Collapse
|
13
|
Yang W, Kim DM, Jiang W, Ai W, Pan Q, Rahman S, Cai JJ, Brashear WA, Sun Y, Guo S. Suppression of FOXO1 attenuates inflamm-aging and improves liver function during aging. Aging Cell 2023; 22:e13968. [PMID: 37602516 PMCID: PMC10577549 DOI: 10.1111/acel.13968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023] Open
Abstract
The liver is a key metabolic organ that maintains whole-body nutrient homeostasis. Aging-induced liver function alterations contribute to systemic susceptibility to aging-related diseases. However, the molecular mechanisms of liver aging remain insufficiently understood. In this study, we performed bulk RNA-Seq and single-cell RNA-Seq analyses to investigate the underlying mechanisms of the aging-induced liver function changes. We found that liver inflammation, glucose intolerance, and liver fat deposition were aggravated in old mice. Aging significantly increased pro-inflammation in hepatic macrophages. Furthermore, we found that Kupffer cells (KCs) were the major driver to induce pro-inflammation in hepatic macrophages during aging. In KCs, aging significantly increased pro-inflammatory levels; in monocyte-derived macrophages (MDMs), aging had a limited effect on pro-inflammation but led to a functional quiescence in antigen presentation and phagosome process. In addition, we identified an aging-responsive KC-specific (ARKC) gene set that potentially mediates aging-induced pro-inflammation in KCs. Interestingly, FOXO1 activity was significantly increased in the liver of old mice. FOXO1 inhibition by AS1842856 significantly alleviated glucose intolerance, hepatic steatosis, and systemic inflammation in old mice. FOXO1 inhibition significantly attenuated aging-induced pro-inflammation in KCs partially through downregulation of ARKC genes. However, FOXO1 inhibition had a limited effect on aging-induced functional quiescence in MDMs. These results indicate that aging induces pro-inflammation in liver mainly through targeting KCs and FOXO1 is a key player in aging-induced pro-inflammation in KCs. Thus, FOXO1 could be a potential therapeutic target for the treatment of age-associated chronic diseases.
Collapse
Affiliation(s)
- Wanbao Yang
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Da Mi Kim
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Shahina Rahman
- Department of StatisticsTexas A&M UniversityCollege StationTexasUSA
| | - James J. Cai
- Department of Veterinary Integrative BiosciencesTexas A&M UniversityCollege StationTexasUSA
| | - Wesley A. Brashear
- High Performance Research ComputingTexas A&M UniversityCollege StationTexasUSA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life SciencesTexas A&M UniversityCollege StationTexasUSA
| |
Collapse
|
14
|
Liu T, Li R, Sun L, Xu Z, Wang S, Zhou J, Wu X, Shi K. Menin orchestrates hepatic glucose and fatty acid uptake via deploying the cellular translocation of SIRT1 and PPARγ. Cell Biosci 2023; 13:175. [PMID: 37740216 PMCID: PMC10517496 DOI: 10.1186/s13578-023-01119-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/30/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Menin is a scaffold protein encoded by the Men1 gene, which interacts with various transcriptional proteins to activate or repress cellular processes and is a key mediator in multiple organs. Both liver-specific and hepatocyte-specific Menin deficiency promotes high-fat diet-induced liver steatosis in mice, as well as insulin resistance and type 2 diabetic phenotype. The potential link between Menin and hepatic metabolism homeostasis may provide new insights into the mechanism of fatty liver disease. RESULTS Disturbance of hepatic Menin expression impacts metabolic pathways associated with non-alcoholic fatty liver disease (NAFLD), including the FoxO signaling pathway, which is similar to that observed in both oleic acid-induced fatty hepatocytes model and biopsied fatty liver tissues, but with elevated hepatic Menin expression and inhibited FABP1. Higher levels of Menin facilitate glucose uptake while restraining fatty acid uptake. Menin targets the expression of FABP3/4/5 and also CD36 or GK, PCK by binding to their promoter regions, while recruiting and deploying the cellular localization of PPARγ and SIRT1 in the nucleus and cytoplasm. Accordingly, Menin binds to PPARγ and/or FoxO1 in hepatocytes, and orchestrates hepatic glucose and fatty acid uptake by recruiting SIRT1. CONCLUSION Menin plays an orchestration role as a transcriptional activator and/or repressor to target downstream gene expression levels involved in hepatic energy uptake by interacting with the cellular energy sensor SIRT1, PPARγ, and/or FoxO1 and deploying their translocations between the cytoplasm and nucleus, thereby maintaining metabolic homeostasis. These findings provide more evidence suggesting Menin could be targeted for the treatment of hepatic steatosis, NAFLD or metabolic dysfunction-associated fatty liver disease (MAFLD), and even other hepatic diseases.
Collapse
Affiliation(s)
- Tingjun Liu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Ranran Li
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Lili Sun
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Zhongjin Xu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Shengxuan Wang
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
| | - Jingxuan Zhou
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Xuanning Wu
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China
| | - Kerong Shi
- Laboratory of Animal Stem Cell and Reprogramming, College of Animal Science and Technology, Shandong Agricultural University, No. 61 Daizong Street, Taian, 271018, Shandong, People's Republic of China.
- Key Laboratory of Animal Bioengineering and Disease Prevention of Shandong Province, Taian, 271018, Shandong, People's Republic of China.
| |
Collapse
|
15
|
Pan Q, Gao M, Kim D, Ai W, Yang W, Jiang W, Brashear W, Dai Y, Li S, Sun Y, Qi Y, Guo S. Hepatocyte FoxO1 Deficiency Protects From Liver Fibrosis via Reducing Inflammation and TGF-β1-mediated HSC Activation. Cell Mol Gastroenterol Hepatol 2023; 17:41-58. [PMID: 37678798 PMCID: PMC10665954 DOI: 10.1016/j.jcmgh.2023.08.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND & AIMS The O-class of the forkhead transcription factor FoxO1 is a crucial factor mediating insulin→PI3K→Akt signaling and governs diverse cellular processes. However, the role of hepatocyte FoxO1 in liver fibrosis has not been well-established. In his study, we investigated the role of hepatocyte FoxO1 in liver fibrosis and uncovered the underlying mechanisms. METHODS Liver fibrosis was established by carbon tetrachloride (CCL4) administration and compared between liver-specific deletion of FoxO1 deletion (F1KO) and control (CNTR) mice. Using genetic and bioinformatic strategies in vitro and in vivo, the role of hepatic FoxO1 in liver fibrosis and associated mechanisms was established. RESULTS Increased FoxO1 expression and FoxO1 signaling activation were observed in CCL4-induced fibrosis. Hepatic FoxO1 deletion largely attenuated CCL4-induced liver injury and fibrosis compared with CNTR mice. F1KO mice showed ameliorated CCL4-induced hepatic inflammation and decreased TGF-β1 mRNA and protein levels compared with those of CNTR mice. In primary hepatocytes, FoxO1 deficiency reduced TGF-β1 expression and secretion. Conditioned medium (CM) collected from wild-type hepatocytes treated with CCL4 activated human HSC cell line (LX-2); such effect was attenuated by FoxO1 deletion in primary hepatocytes or neutralization of TGF-β1 in the CM using TGF-β1 antibody. Hepatic FoxO1 overexpression in CNTR mice promoted CCL4-induced HSC activation; such effect was blocked in L-TGF-β1KO mice. CONCLUSIONS Hepatic FoxO1 mediates CCL4-inducled liver fibrosis via upregulating hepatocyte TGF-β1 expression, stimulating hepatic inflammation and TGF-β1-mediated HSC activation. Hepatic FoxO1 may be a therapeutic target for prevention and treatment of liver fibrosis.
Collapse
Affiliation(s)
- Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Mingming Gao
- Department of Pharmacology, School of Basic Medical Science, North China University of Science and Technology. Tangshan, China
| | - DaMi Kim
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Wesley Brashear
- High Performance Research Computing, Texas A&M University, College Station, Texas
| | - Yujiao Dai
- Department of Pharmacology, School of Basic Medical Science, North China University of Science and Technology. Tangshan, China
| | - Sha Li
- Department of Pharmacology, School of Basic Medical Science, North China University of Science and Technology. Tangshan, China
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas
| | - Yajuan Qi
- Department of Pharmacology, School of Basic Medical Science, North China University of Science and Technology. Tangshan, China.
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas.
| |
Collapse
|
16
|
Chen Y, Pan Q, Liao W, Ai W, Yang S, Guo S. Transcription Factor Forkhead Box O1 Mediates Transforming Growth Factor-β1-Induced Apoptosis in Hepatocytes. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1143-1155. [PMID: 37263346 PMCID: PMC10477955 DOI: 10.1016/j.ajpath.2023.05.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/27/2023] [Accepted: 05/18/2023] [Indexed: 06/03/2023]
Abstract
Dysregulation of hepatocyte apoptosis is associated with several types of chronic liver diseases. Transforming growth factor-β1 (TGF-β1) is a well-known pro-apoptotic factor in the liver, which constitutes a receptor complex composed of TGF-β receptor I and II, along with transcription factor Smad proteins. As a member of the forkhead box O (Foxo) class of transcription factors, Foxo1 is a predominant regulator of hepatic glucose production and apoptosis. This study investigated the potential relationship between TGF-β1 signaling and Foxo1 in control of apoptosis in hepatocytes. TGF-β1 induced hepatocyte apoptosis in a Foxo1-dependent manner in hepatocytes isolated from both wild-type and liver-specific Foxo1 knockout mice. TGF-β1 activated protein kinase A through TGF-β receptor I-Smad3, followed by phosphorylation of Foxo1 at Ser273 in promotion of apoptosis in hepatocytes. Moreover, Smad3 overexpression in the liver of mice promoted the levels of phosphorylated Foxo1-S273, total Foxo1, and a Foxo1-target pro-apoptotic gene Bim, which eventually resulted in hepatocyte apoptosis. The study further demonstrated a crucial role of Foxo1-S273 phosphorylation in the pro-apoptotic effect of TGF-β1 by using hepatocytes isolated from Foxo1-S273A/A knock-in mice, in which the phosphorylation of Foxo1-S273 was disrupted. Taken together, this study established a novel role of TGF-β1→protein kinase A→Foxo1 signaling cascades in control of hepatocyte survival.
Collapse
Affiliation(s)
- Yunmei Chen
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Quan Pan
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Wang Liao
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Weiqi Ai
- Department of Nutrition, Texas A&M University, College Station, Texas
| | - Sijun Yang
- Institute of Animal Model for Human Disease, Wuhan University, Wuhan, China
| | - Shaodong Guo
- Department of Nutrition, Texas A&M University, College Station, Texas.
| |
Collapse
|
17
|
Pan Q, Ai W, Chen Y, Kim DM, Shen Z, Yang W, Jiang W, Sun Y, Safe S, Guo S. Reciprocal Regulation of Hepatic TGF-β1 and Foxo1 Controls Gluconeogenesis and Energy Expenditure. Diabetes 2023; 72:1193-1206. [PMID: 37343276 PMCID: PMC10450826 DOI: 10.2337/db23-0180] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/13/2023] [Indexed: 06/23/2023]
Abstract
Obesity and insulin resistance are risk factors for the pathogenesis of type 2 diabetes (T2D). Here, we report that hepatic TGF-β1 expression positively correlates with obesity and insulin resistance in mice and humans. Hepatic TGF-β1 deficiency decreased blood glucose levels in lean mice and improved glucose and energy dysregulations in diet-induced obese (DIO) mice and diabetic mice. Conversely, overexpression of TGF-β1 in the liver exacerbated metabolic dysfunctions in DIO mice. Mechanistically, hepatic TGF-β1 and Foxo1 are reciprocally regulated: fasting or insulin resistance caused Foxo1 activation, increasing TGF-β1 expression, which, in turn, activated protein kinase A, stimulating Foxo1-S273 phosphorylation to promote Foxo1-mediated gluconeogenesis. Disruption of TGF-β1→Foxo1→TGF-β1 looping by deleting TGF-β1 receptor II in the liver or by blocking Foxo1-S273 phosphorylation ameliorated hyperglycemia and improved energy metabolism in adipose tissues. Taken together, our studies reveal that hepatic TGF-β1→Foxo1→TGF-β1 looping could be a potential therapeutic target for prevention and treatment of obesity and T2D. ARTICLE HIGHLIGHTS Hepatic TGF-β1 levels are increased in obese humans and mice. Hepatic TGF-β1 maintains glucose homeostasis in lean mice and causes glucose and energy dysregulations in obese and diabetic mice. Hepatic TGF-β1 exerts an autocrine effect to promote hepatic gluconeogenesis via cAMP-dependent protein kinase-mediated Foxo1 phosphorylation at serine 273, endocrine effects on brown adipose tissue action, and inguinal white adipose tissue browning (beige fat), causing energy imbalance in obese and insulin-resistant mice. TGF-β1→Foxo1→TGF-β1 looping in hepatocytes plays a critical role in controlling glucose and energy metabolism in health and disease.
Collapse
Affiliation(s)
- Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yunmei Chen
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Da Mi Kim
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Zheng Shen
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Stephen Safe
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| |
Collapse
|
18
|
Yang W, Liao W, Li X, Ai W, Pan Q, Shen Z, Jiang W, Guo S. Hepatic p38α MAPK controls gluconeogenesis via FOXO1 phosphorylation at S273 during glucagon signalling in mice. Diabetologia 2023:10.1007/s00125-023-05916-5. [PMID: 37202506 DOI: 10.1007/s00125-023-05916-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 02/09/2023] [Indexed: 05/20/2023]
Abstract
AIMS/HYPOTHESIS Hyperglucagonaemia-stimulated hepatic glucose production (HGP) contributes to hyperglycaemia during type 2 diabetes. A better understanding of glucagon action is important to enable efficient therapies to be developed for the treatment of diabetes. Here, we aimed to investigate the role of p38 MAPK family members in glucagon-induced HGP and determine the underlying mechanisms by which p38 MAPK regulates glucagon action. METHODS p38α, β, γ and δ MAPK siRNAs were transfected into primary hepatocytes, followed by measurement of glucagon-induced HGP. Adeno-associated virus serotype 8 carrying p38α MAPK short hairpin RNA (shRNA) was injected into liver-specific Foxo1 knockout, liver-specific Irs1/Irs2 double knockout and Foxo1S273D knockin mice. Foxo1S273A knockin mice were fed a high-fat diet for 10 weeks. Pyruvate tolerance tests, glucose tolerance tests, glucagon tolerance tests and insulin tolerance tests were carried out in mice, liver gene expression profiles were analysed and serum triglyceride, insulin and cholesterol levels were measured. Phosphorylation of forkhead box protein O1 (FOXO1) by p38α MAPK in vitro was analysed by LC-MS. RESULTS We found that p38α MAPK, but not the other p38 isoforms, stimulates FOXO1-S273 phosphorylation and increases FOXO1 protein stability, promoting HGP in response to glucagon stimulation. In hepatocytes and mouse models, inhibition of p38α MAPK blocked FOXO1-S273 phosphorylation, decreased FOXO1 levels and significantly impaired glucagon- and fasting-induced HGP. However, the effect of p38α MAPK inhibition on HGP was abolished by FOXO1 deficiency or a Foxo1 point mutation at position 273 from serine to aspartic acid (Foxo1S273D) in both hepatocytes and mice. Moreover, an alanine mutation at position 273 (Foxo1S273A) decreased glucose production, improved glucose tolerance and increased insulin sensitivity in diet-induced obese mice. Finally, we found that glucagon activates p38α through exchange protein activated by cAMP 2 (EPAC2) signalling in hepatocytes. CONCLUSIONS/INTERPRETATION This study found that p38α MAPK stimulates FOXO1-S273 phosphorylation to mediate the action of glucagon on glucose homeostasis in both health and disease. The glucagon-induced EPAC2-p38α MAPK-pFOXO1-S273 signalling pathway is a potential therapeutic target for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
| | - Wang Liao
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
| | - Xiaopeng Li
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
| | - Zheng Shen
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
19
|
Liang M, Wang L, Wang W. The 15-hydroxyprostaglandin dehydrogenase inhibitor SW033291 ameliorates abnormal hepatic glucose metabolism through PGE 2-EP4 receptor-AKT signaling in a type 2 diabetes mellitus mouse model. Cell Signal 2023; 108:110707. [PMID: 37164143 DOI: 10.1016/j.cellsig.2023.110707] [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: 02/24/2023] [Revised: 04/21/2023] [Accepted: 05/03/2023] [Indexed: 05/12/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is associated with high rates of morbidity and mortality worldwide. Prostaglandin E2 (PGE2) is a lipid signaling molecule that can ameliorate the symptoms of some metabolic diseases, including T2DM, and improve tissue repair and regeneration. Although SW033291 can increase PGE2 levels through its action as a small molecule inhibitor of the PGE2-degrading enzyme 15-hydroxyprostaglandin dehydrogenase, its effects on T2DM remain unclear. In the present study, we evaluated whether SW033291 treatment exerts a protective effect against T2DM and explored the underlying mechanisms. A T2DM mouse model was established using a high-fat diet combined with streptozotocin treatment. Palmitic acid-treated LO2 cells were used as an insulin-resistant cell model. SW033291 treatment reduced body weight and fasting blood glucose levels as well as serum triglyceride, total cholesterol, and low-density lipoprotein cholesterol levels in vivo. In addition to ameliorating glucose and insulin tolerance, SW033291 treatment reversed the T2DM-induced decrease in glycogen synthesis and increase in gluconeogenesis in the liver. Furthermore, SW033291 administration increased hepatic glycogen synthase kinase 3 beta (GSK3β) phosphorylation levels to promote glycogen synthesis. SW033291 treatment also inhibited gluconeogenesis by upregulating AKT serine/threonine kinase (AKT) and forkhead box O1 (FOXO1) phosphorylation and reducing glucose-6-phosphatase and phosphoenolpyruvate carboxykinase 1 expression in the livers of T2DM model mice. Additionally, SW033291 treatment improved abnormal hepatic glucose metabolism through the PGE2-EP4 receptor-AKT-GSK3β/FOXO1 signaling pathway in vitro. These results suggest a novel role of SW033291 in improving T2DM and support its potential as a novel therapeutic agent.
Collapse
Affiliation(s)
- Mingjie Liang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China; Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou, Guangdong Province, China; Key Laboratory of Glucolipid Metabolic Diseases, Ministry of Education, Guangzhou, Guangdong Province, China; Guangdong Provincial TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Lexun Wang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China; Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou, Guangdong Province, China; Key Laboratory of Glucolipid Metabolic Diseases, Ministry of Education, Guangzhou, Guangdong Province, China; Guangdong Provincial TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Weixuan Wang
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China; Guangdong Provincial Research Center of Integration of Traditional Chinese Medicine and Western Medicine in Metabolic Diseases, Guangzhou, Guangdong Province, China; Key Laboratory of Glucolipid Metabolic Diseases, Ministry of Education, Guangzhou, Guangdong Province, China; Guangdong Provincial TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
20
|
Azarova I, Polonikov A, Klyosova E. Molecular Genetics of Abnormal Redox Homeostasis in Type 2 Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24054738. [PMID: 36902173 PMCID: PMC10003739 DOI: 10.3390/ijms24054738] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Numerous studies have shown that oxidative stress resulting from an imbalance between the production of free radicals and their neutralization by antioxidant enzymes is one of the major pathological disorders underlying the development and progression of type 2 diabetes (T2D). The present review summarizes the current state of the art advances in understanding the role of abnormal redox homeostasis in the molecular mechanisms of T2D and provides comprehensive information on the characteristics and biological functions of antioxidant and oxidative enzymes, as well as discusses genetic studies conducted so far in order to investigate the contribution of polymorphisms in genes encoding redox state-regulating enzymes to the disease pathogenesis.
Collapse
Affiliation(s)
- Iuliia Azarova
- Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| | - Alexey Polonikov
- Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, 305041 Kursk, Russia
- Correspondence:
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya Street, 305041 Kursk, Russia
| |
Collapse
|
21
|
Wu LX, Xu YC, Pantopoulos K, Tan XY, Wei XL, Zheng H, Luo Z. Glycophagy mediated glucose-induced changes of hepatic glycogen metabolism via OGT1-AKT1-FOXO1Ser238 pathway. J Nutr Biochem 2023; 117:109337. [PMID: 36990368 DOI: 10.1016/j.jnutbio.2023.109337] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 03/12/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023]
Abstract
Glycophagy is the autophagy degradation of glycogen. However, the regulatory mechanisms for glycophagy and glucose metabolism remain unexplored. Herein, we demonstrated that high-carbohydrate diet (HCD) and high glucose (HG) incubation induced glycogen accumulation, AKT1 expression and AKT1-dependent phosphorylation of forkhead transcription factor O1 (FOXO1) at Ser238 in the liver tissues and hepatocytes. The glucose-induced FOXO1 phosphorylation at Ser238 prevents FOXO1 entry into the nucleus and the recruitment to the gabarapl1 promoter, reduces the gabarapl1 promoter activity, and inhibits glycophagy and glucose production. The glucose-dependent O-GlcNAcylation of AKT1 by OGT1 enhances the stability of AKT1 protein and promotes its binding with FOXO1. Moreover, the glycosylation of AKT1 is crucial for promoting FOXO1 nuclear translocation and inhibiting glycophagy. Our studies elucidate a novel mechanism for glycophagy inhibition by high carbohydrate and glucose via OGT1-AKT1-FOXO1Ser238 pathway in the liver tissues and hepatocytes, which provides critical insights into potential intervention strategies for glycogen storage disorders in vertebrates, as well as human beings.
Collapse
|
22
|
Ramatchandirin B, Pearah A, He L. Regulation of Liver Glucose and Lipid Metabolism by Transcriptional Factors and Coactivators. Life (Basel) 2023; 13:life13020515. [PMID: 36836874 PMCID: PMC9962321 DOI: 10.3390/life13020515] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) worldwide is on the rise and NAFLD is becoming the most common cause of chronic liver disease. In the USA, NAFLD affects over 30% of the population, with similar occurrence rates reported from Europe and Asia. This is due to the global increase in obesity and type 2 diabetes mellitus (T2DM) because patients with obesity and T2DM commonly have NAFLD, and patients with NAFLD are often obese and have T2DM with insulin resistance and dyslipidemia as well as hypertriglyceridemia. Excessive accumulation of triglycerides is a hallmark of NAFLD and NAFLD is now recognized as the liver disease component of metabolic syndrome. Liver glucose and lipid metabolisms are intertwined and carbon flux can be used to generate glucose or lipids; therefore, in this review we discuss the important transcription factors and coactivators that regulate glucose and lipid metabolism.
Collapse
Affiliation(s)
| | - Alexia Pearah
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 600 N. Wolfe St, Baltimore, MD 21287, USA
- Correspondence: ; Tel.: +1-410-502-5765; Fax: +1-410-502-5779
| |
Collapse
|
23
|
Wang K, Cui X, Li F, Xia L, Wei T, Liu J, Fu W, Yang J, Hong T, Wei R. Glucagon receptor blockage inhibits β-cell dedifferentiation through FoxO1. Am J Physiol Endocrinol Metab 2023; 324:E97-E113. [PMID: 36383639 DOI: 10.1152/ajpendo.00101.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Glucagon-secreting pancreatic α-cells play pivotal roles in the development of diabetes. Glucagon promotes insulin secretion from β-cells. However, the long-term effect of glucagon on the function and phenotype of β-cells had remained elusive. In this study, we found that long-term glucagon intervention or glucagon intervention with the presence of palmitic acid downregulated β-cell-specific markers and inhibited insulin secretion in cultured β-cells. These results suggested that glucagon induced β-cell dedifferentiation under pathological conditions. Glucagon blockage by a glucagon receptor (GCGR) monoclonal antibody (mAb) attenuated glucagon-induced β-cell dedifferentiation. In primary islets, GCGR mAb treatment upregulated β-cell-specific markers and increased insulin content, suggesting that blockage of endogenous glucagon-GCGR signaling inhibited β-cell dedifferentiation. To investigate the possible mechanism, we found that glucagon decreased FoxO1 expression. FoxO1 inhibitor mimicked the effect of glucagon, whereas FoxO1 overexpression reversed the glucagon-induced β-cell dedifferentiation. In db/db mice and β-cell lineage-tracing diabetic mice, GCGR mAb lowered glucose level, upregulated plasma insulin level, increased β-cell area, and inhibited β-cell dedifferentiation. In aged β-cell-specific FoxO1 knockout mice (with the blood glucose level elevated as a diabetic model), the glucose-lowering effect of GCGR mAb was attenuated and the plasma insulin level, β-cell area, and β-cell dedifferentiation were not affected by GCGR mAb. Our results proved that glucagon induced β-cell dedifferentiation under pathological conditions, and the effect was partially mediated by FoxO1. Our study reveals a novel cross talk between α- and β-cells and is helpful to understand the pathophysiology of diabetes and discover new targets for diabetes treatment.NEW & NOTEWORTHY Glucagon-secreting pancreatic α-cells can interact with β-cells. However, the long-term effect of glucagon on the function and phenotype of β-cells has remained elusive. Our new finding shows that long-term glucagon induces β-cell dedifferentiation in cultured β-cells. FoxO1 inhibitor mimicks whereas glucagon signaling blockage by GCGR mAb reverses the effect of glucagon. In type 2 diabetic mice, GCGR mAb increases β-cell area, improves β-cell function, and inhibits β-cell dedifferentiation, and the effect is partially mediated by FoxO1.
Collapse
Affiliation(s)
- Kangli Wang
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
| | - Xiaona Cui
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
| | - Fei Li
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
| | - Li Xia
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
| | - Tianjiao Wei
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
| | - Junling Liu
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
| | - Wei Fu
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
| | - Jin Yang
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Tianpei Hong
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| | - Rui Wei
- Department of Endocrinology and Metabolism, https://ror.org/04wwqze12Peking University Third Hospital, Beijing, China
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, China
| |
Collapse
|
24
|
Protein Targeting to Glycogen (PTG): A Promising Player in Glucose and Lipid Metabolism. Biomolecules 2022; 12:biom12121755. [PMID: 36551183 PMCID: PMC9775135 DOI: 10.3390/biom12121755] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Protein phosphorylation and dephosphorylation are widely considered to be the key regulatory factors of cell function, and are often referred to as "molecular switches" in the regulation of cell metabolic processes. A large number of studies have shown that the phosphorylation/dephosphorylation of related signal molecules plays a key role in the regulation of liver glucose and lipid metabolism. As a new therapeutic strategy for metabolic diseases, the potential of using inhibitor-based therapies to fight diabetes has gained scientific momentum. PTG, a protein phosphatase, also known as glycogen targeting protein, is a member of the protein phosphatase 1 (PP1) family. It can play a role by catalyzing the dephosphorylation of phosphorylated protein molecules, especially regulating many aspects of glucose and lipid metabolism. In this review, we briefly summarize the role of PTG in glucose and lipid metabolism, and update its role in metabolic regulation, with special attention to glucose homeostasis and lipid metabolism.
Collapse
|
25
|
Zhou P, Deng F, Yang Z, Cao C, Zhao H, Liu F, Zhong K, Fu L, Peng T, Sun D, Liu H, Li R, Yu Y. Ginsenoside Rb1 inhibits oxidative stress-induced ovarian granulosa cell injury through Akt-FoxO1 interaction. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2301-2315. [PMID: 35661967 DOI: 10.1007/s11427-021-2080-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/28/2022] [Indexed: 06/15/2023]
Abstract
Ginsenoside Rb1 shows a strong antioxidant effect and has potential activation effects on Akt. The aim of the present study was to investigate the protective effect of Rb1 on age-related ovarian granulosa cell injury. Ovarian granulosa cells (GCs) were obtained from 50 young women (≤30 years) and 50 aged women (≥38 years) at an IVF center. Young and aged ICR mice were administered with or without Rb1 (10 mg kg-1, i.p.) for 2 weeks. The protective effects of Rb1 were investigated and the role of Rb1 on the modulation of Akt-FoxO1 interaction was determined with immunofluorescence, Western blotting, immunoprecipitation, siRNA silencing and pharmacological inhibitor. Rb1 effectively decreased LDH and MDA, and reversed the apoptotic-related protein levels in hGL cells from old patients. Similar results were found in mice. In addition, the mitochondrial membrane potential was restored and the overaccumulation of ROS was reversed by Rb1. Rb1 preserved peroxide-impaired Akt activation, to some extent, by increasing phosphorylation at Ser473. Rb1 also facilitated p-Akt binding to FoxO1 and promoted the phosphorylation of FoxO1. SiRNA silencing of Akt, Akt inhibitor LY294002, and FoxO1 inhibitor AS1842856 attenuated the effects of Rb1. Ginsenoside Rb1 inhibits age-related GCs oxidative damage by activating Akt phosphorylation at Ser473 and by further interaction with FoxO1.
Collapse
Affiliation(s)
- Ping Zhou
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Feng Deng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Zi Yang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Canhui Cao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Hongcui Zhao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Fenting Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Ke Zhong
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Lin Fu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Tianliu Peng
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Di Sun
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Hui Liu
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China
| | - Rong Li
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China
| | - Yang Yu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, 100191, China.
- Clinical Stem Cell Research Center, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
26
|
Transcriptional regulation of autophagy in aging. CURRENT OPINION IN PHYSIOLOGY 2022. [DOI: 10.1016/j.cophys.2022.100591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
27
|
Zhuang L, Li C, Hu X, Yang Q, Pei X, Jin G. High expression of P4HA3 in obesity: a potential therapeutic target for type 2 diabetes. Braz J Med Biol Res 2022; 55:e11741. [PMID: 35976267 PMCID: PMC9377532 DOI: 10.1590/1414-431x2022e11741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/19/2022] [Indexed: 11/22/2022] Open
Abstract
The aims of the present study were to evaluate the expression of prolyl 4-hydroxylase subunit alpha 3 (P4HA3) in adipocytes and adipose tissue and to explore its effect on obesity and type 2 diabetes mellitus (T2DM). We initially demonstrated that P4HA3 was significantly upregulated in the subcutaneous adipose tissue of obesity and T2DM patients, and its functional roles in adipocyte differentiation and insulin resistance were investigated using in vitro and in vivo models. The knockdown of P4HA3 inhibited adipocyte differentiation and improved insulin resistance in 3T3-L1 cells. In C57BL/6J db/db mice fed with a high fat diet (HFD), silencing P4HA3 significantly decreased fasting blood glucose and triglycerides (TG) levels, with concomitant decrease of body weight and adipose tissue weight. Further analysis showed that P4HA3 knockdown was correlated with the augmented IRS-1/PI3K/Akt/FoxO1 signaling pathway in the adipose and hepatic tissues of obese mice, which could improve hepatic glucose homeostasis and steatosis of mice. Together, our study suggested that the dysregulation of P4HA3 may contribute to the development of obesity and T2DM.
Collapse
Affiliation(s)
- Langen Zhuang
- Department of Endocrinology, The First Affiliated Hospital of
Bengbu Medical College, Bengbu, Anhui, China
| | - Can Li
- Shangyi Health Check-up Centre, Zibo, Shandong, China
| | - Xiaolei Hu
- Department of Endocrinology, The First Affiliated Hospital of
Bengbu Medical College, Bengbu, Anhui, China
| | - Qingqing Yang
- Department of Endocrinology, The First Affiliated Hospital of
Bengbu Medical College, Bengbu, Anhui, China
| | - Xiaoyan Pei
- Department of Endocrinology, The First Affiliated Hospital of
Bengbu Medical College, Bengbu, Anhui, China
| | - Guoxi Jin
- Department of Endocrinology, The First Affiliated Hospital of
Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
28
|
Cao X, Liao W, Wang S. Food protein-derived bioactive peptides for the management of nutrition related chronic diseases. ADVANCES IN FOOD AND NUTRITION RESEARCH 2022; 101:277-307. [PMID: 35940708 DOI: 10.1016/bs.afnr.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Dietary intervention via modifications of dietary pattern or supplementations of naturally derived bioactive compounds has been considered as an efficient approach in management of nutrition related chronic diseases. Food protein-derived bioactive peptide is representative of natural compounds which show the potential to prevent or mitigate nutrition related chronic diseases. In the past decades, substantial research has been conducted concentrating on the characterization, bioavailability, and activity assessment of bioactive peptides. Although various activities of bioactive peptides have been reported, the activity testes of most peptides were only conducted in cells and animal models. Some clinical trials of bioactive peptides were also reported but only limited to antihypertensive peptides, antidiabetic peptides and peptides modulating blood lipid profile. Hereby, clinical evidence of bioactive peptides in management of nutrition-related chronic diseases is summarized in this chapter, which aims at providing implications for the clinical studies of bioactive peptides in the future.
Collapse
Affiliation(s)
- Xinyi Cao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| | - Wang Liao
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, China.
| | - Shaokang Wang
- Key Laboratory of Environmental Medicine and Engineering of Ministry of Education, and Department of Nutrition and Food Hygiene, School of Public Health, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
29
|
Kamiya T, Yamaguchi Y, Oka M, Hara H. Combined action of FOXO1 and superoxide dismutase 3 promotes MDA-MB-231 cell migration. Free Radic Res 2022; 56:106-114. [PMID: 35271779 DOI: 10.1080/10715762.2022.2049770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Superoxide dismutase 3 (SOD3), one of SOD isozymes, maintains extracellular redox homeostasis through the dismutation reaction of superoxide. Loss of SOD3 in tumor cells induces oxidative stress and exacerbates tumor progression; however, interestingly, overexpression of SOD3 also promotes cell proliferation through the production of hydrogen peroxide. In this study, we investigated the functional role of SOD3 in human breast cancer MDA-MB-231 cell migration and the molecular mechanisms involved in high expression of SOD3 in MDA-MB-231 cells and human monocytic THP-1 cells. The level of histone H3 trimethylation at lysine 27 (H3K27me3), a marker of gene silencing, was decreased in 12-O-tetra-decanoylphorbol-13-acetate (TPA)-treated THP-1 cells. Also, that reduction was observed within the SOD3 promoter region. We then investigated the involvement of H3K27 demethylase JMJD3 in SOD3 induction. The induction of SOD3 and the reduction of H3K27me3 were inhibited in the presence of JMJD3 inhibitor, GSK-J4. Additionally, it was first determined that the knockdown of the transcription factor forkhead box O1 (FOXO1) significantly suppressed TPA-elicited SOD3 induction. FOXO1-mediated SOD3 downregulation was also observed in MDA-MB-231 cells, and knockdown of FOXO1 and SOD3 suppressed cell migration. Our results provide a novel insight into epigenetic regulation of SOD3 expression in tumor-associated cells, and high expression of FOXO1 and SOD3 would participate in the migration of MDA-MB-231 cells.
Collapse
Affiliation(s)
- Tetsuro Kamiya
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Yuji Yamaguchi
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Manami Oka
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Hirokazu Hara
- Laboratory of Clinical Pharmaceutics, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
30
|
Wang S, Liu T, Sun L, Du H, Xu Z, Li R, Yu Y, Mao Y, Shi K. Menin regulates lipid deposition in mouse hepatocytes via interacting with transcription factor FoxO1. Mol Cell Biochem 2022; 477:1555-1568. [PMID: 35182330 DOI: 10.1007/s11010-022-04392-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 02/10/2022] [Indexed: 11/25/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is rapidly being recognized as the leading cause of chronic liver disease worldwide. Men1, encoding protein of menin, is a key causative gene of multiple endocrine neoplasia type 1 syndrome including pancreatic tumor. It is known that insulin that secretes by endocrine tissue pancreatic islets plays a critical role in hepatic metabolism. Mouse model of hemizygous deletion of Men1 was shown to have severe hepatic metabolism disorders. However, the molecular function of menin on lipid deposition in hepatocytes needs to be further studied. Transcriptome sequencing does show that expression suppression of Men1 in mouse hepatocytes widely affect signaling pathways involved in hepatic metabolism, such as fatty acid metabolism, insulin response, glucose metabolism and inflammation. Further molecular studies indicates that menin overexpression inhibits expressions of the fat synthesis genes Srebp-1c, Fas, and Acc1, the fat differentiation genes Pparγ1 and Pparγ2, and the fat transport gene Cd36, thereby inhibiting the fat accumulation in hepatocytes. The biological process of menin regulating hepatic lipid metabolism was accomplished by interacting with the transcription factor FoxO1, which is also found to be critical for lipid metabolism. Moreover, menin responds to insulin in hepatocytes and mediates its regulatory effect on hepatic metabolism. Our findings suggest that menin is a crucial mediation factor in regulating the hepatic fat deposition, suggesting it could be a potential important therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Shengxuan Wang
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Tingjun Liu
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Lili Sun
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Hongxia Du
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Zhongjin Xu
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Ranran Li
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China
| | - Ying Yu
- National Engineering Laboratory for Animal Breeding, MOA Key Laboratory of Animal Genetics and Breeding, Department of Animal Genetics and Breeding, China Agricultural University, Beijing, 100093, China
| | - Yongjiang Mao
- Key Laboratory of Animal Genetics & Breeding and Molecular Design of Jiangsu Province, Yangzhou University, Yangzhou, 225009, China
| | - Kerong Shi
- College of Animal Science and Technology, Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, Shandong Agricultural University, No. 61 Daizong Street, Taian, Shandong, 271018, China.
| |
Collapse
|
31
|
Khan MF, Mathur A, Pandey VK, Kakkar P. Endoplasmic reticulum stress-dependent activation of TRB3-FoxO1 signaling pathway exacerbates hyperglycemic nephrotoxicity: Protection accorded by Naringenin. Eur J Pharmacol 2022; 917:174745. [PMID: 34998792 DOI: 10.1016/j.ejphar.2022.174745] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/07/2021] [Accepted: 01/03/2022] [Indexed: 12/22/2022]
Abstract
Endoplasmic reticulum (ER) dysfunction contributes greatly to the pathophysiology of hyperglycemic nephrotoxicity. This study unravels the critical role of Tribbles 3 (TRB3)-Forkhead box O1 (FoxO1) signaling pathway during hyperglycemic renal toxicity. It also uncovers the novel role of Naringenin, a flavanone, in regulating ER stress in proximal tubular cells, NRK 52E, and kidneys of streptozotocin/nicotinamide induced experimental diabetic Wistar rats. Results demonstrate that expression of ER stress marker proteins including phosphorylated protein kinase ER like kinase (p-PERK), phosphorylated eukaryotic Initiation Factor 2α (p-eIF2α), X Box Binding Protein 1 spliced (XBP1s), Activating Transcription Factor 4 (ATF4) and C/EBP Homologous Protein (CHOP) were upregulated in diabetic kidneys indicating the activation of ER stress response due to nephrotoxicity. Treatment with Naringenin reduced the expression of TRB3, an ER stress-inducible pseudokinase, both in vitro and in vivo. Gene silencing of TRB3 enhanced Akt and FoxO1 phosphorylation and alleviated FoxO1 mediated apoptosis during hyperglycemic nephrotoxicity. Notably, TRB3 gene silencing effects were comparable to the response with Naringenin treatment. Prevention of nuclear colocalization of ATF4 and CHOP in Naringenin treated cells was evident. Naringenin also reduced insulin resistance, apoptosis and glycogen accumulation along with enhancement of glucose tolerance in diabetic rats. Prevention of ultrastructural aberrations in the ER of hyperglycemic renal cells by Naringenin confirmed its anti-ER stress effects. These findings affirm that activation of TRB3-FoxO1 signaling is critical in the pathogenesis of hyperglycemia-induced renal toxicity and protective effect of Naringenin via modulation of ER stress may be exploited as a novel approach for its management.
Collapse
Affiliation(s)
- Mohammad Fareed Khan
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Alpana Mathur
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India
| | - Vivek Kumar Pandey
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Poonam Kakkar
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
32
|
Emergent Roles of Circular RNAs in Metabolism and Metabolic Disorders. Int J Mol Sci 2022; 23:ijms23031032. [PMID: 35162956 PMCID: PMC8834750 DOI: 10.3390/ijms23031032] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 12/24/2022] Open
Abstract
Circular RNAs (circRNAs) are an emerging group of long non-coding RNAs (lncRNAs) and have attracted attention again according to the progress in high-throughput sequencing in recent years. circRNAs are genome transcripts produced from pre-messenger (m)RNA regions in a specific process called “back-splicing,” which forms covalently closed continuous loops. Due to their lack of a 5’ cap and 3’ poly-adenylated tails, circRNAs are remarkably more stable than linear RNAs. Functionally, circRNAs can endogenously sponge to microRNAs, interact with RNA-binding proteins (RBPs), or translate themselves. Moreover, circRNAs can be expressed in cell type- or tissue-specific expression patterns. Therefore, they are proposed to play essential roles in fine-tuning our body’s homeostasis by regulating transcription and translation processes. Indeed, there has been accumulating emergent evidence showing that dysregulation of circRNAs can lead to metabolic disorders. This study explored the current knowledge of circRNAs that regulate molecular processes associated with glucose and lipid homeostasis and related pathogeneses of metabolic disorders. We also suggest the potential role of circRNAs as disease biomarkers and therapeutic targets.
Collapse
|
33
|
Nathanael J, Suardana P, Vianney YM, Dwi Putra SE. The role of FoxO1 and its modulation with small molecules in the development of diabetes mellitus: A review. Chem Biol Drug Des 2021; 99:344-361. [PMID: 34862852 DOI: 10.1111/cbdd.13989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/11/2021] [Accepted: 11/21/2021] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus type 2 (T2D) is one of the metabolic disorders suffered by a global human being. Certain factors, such as lifestyle and heredity, can increase a person's tendency for T2D. Various genes and proteins play a role in the development of insulin resistance and ultimately diabetes in which one central protein that is discussed in this review is FoxO1. In this review, we regard FoxO1 activation as detrimental, promote high plasma glucose level, and induce insulin resistance. Indeed, many contrasting studies arise since FoxO1 is an important protein to alleviate oxidative stress and promote cell survival, for example, also by preventing hyperglycemic-induced cell death. Inter-relation to PPARG, another important protein in metabolism, is also discussed. Ultimately, we discussed contrasting approaches of targeting FoxO1 to combat diabetes mellitus by small molecules.
Collapse
Affiliation(s)
- Joshua Nathanael
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Putu Suardana
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Yoanes Maria Vianney
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Sulistyo Emantoko Dwi Putra
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| |
Collapse
|
34
|
How pervasive are post-translational and -transcriptional modifications? Trends Cell Biol 2021; 32:475-478. [PMID: 34863586 DOI: 10.1016/j.tcb.2021.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/06/2021] [Accepted: 11/10/2021] [Indexed: 12/19/2022]
Abstract
Cells use post-translational and post-transcription modifications as crucial mechanisms to maintain homeostasis and regulate gene transcription. Recent discoveries demonstrate that these modifications are more pervasive and important than scientists previously posited. Here, we discuss their importance and provide insight to stimulate new research into these modifications.
Collapse
|
35
|
Saydmohammed M, Jha A, Mahajan V, Gavlock D, Shun TY, DeBiasio R, Lefever D, Li X, Reese C, Kershaw EE, Yechoor V, Behari J, Soto-Gutierrez A, Vernetti L, Stern A, Gough A, Miedel MT, Lansing Taylor D. Quantifying the progression of non-alcoholic fatty liver disease in human biomimetic liver microphysiology systems with fluorescent protein biosensors. Exp Biol Med (Maywood) 2021; 246:2420-2441. [PMID: 33957803 PMCID: PMC8606957 DOI: 10.1177/15353702211009228] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 03/23/2021] [Indexed: 12/12/2022] Open
Abstract
Metabolic syndrome is a complex disease that involves multiple organ systems including a critical role for the liver. Non-alcoholic fatty liver disease (NAFLD) is a key component of the metabolic syndrome and fatty liver is linked to a range of metabolic dysfunctions that occur in approximately 25% of the population. A panel of experts recently agreed that the acronym, NAFLD, did not properly characterize this heterogeneous disease given the associated metabolic abnormalities such as type 2 diabetes mellitus (T2D), obesity, and hypertension. Therefore, metabolic dysfunction-associated fatty liver disease (MAFLD) has been proposed as the new term to cover the heterogeneity identified in the NAFLD patient population. Although many rodent models of NAFLD/NASH have been developed, they do not recapitulate the full disease spectrum in patients. Therefore, a platform has evolved initially focused on human biomimetic liver microphysiology systems that integrates fluorescent protein biosensors along with other key metrics, the microphysiology systems database, and quantitative systems pharmacology. Quantitative systems pharmacology is being applied to investigate the mechanisms of NAFLD/MAFLD progression to select molecular targets for fluorescent protein biosensors, to integrate computational and experimental methods to predict drugs for repurposing, and to facilitate novel drug development. Fluorescent protein biosensors are critical components of the platform since they enable monitoring of the pathophysiology of disease progression by defining and quantifying the temporal and spatial dynamics of protein functions in the biosensor cells, and serve as minimally invasive biomarkers of the physiological state of the microphysiology system experimental disease models. Here, we summarize the progress in developing human microphysiology system disease models of NAFLD/MAFLD from several laboratories, developing fluorescent protein biosensors to monitor and to measure NAFLD/MAFLD disease progression and implementation of quantitative systems pharmacology with the goal of repurposing drugs and guiding the creation of novel therapeutics.
Collapse
Affiliation(s)
- Manush Saydmohammed
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Anupma Jha
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Vineet Mahajan
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Dillon Gavlock
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Tong Ying Shun
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Richard DeBiasio
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Daniel Lefever
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Xiang Li
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Celeste Reese
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Erin E Kershaw
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Vijay Yechoor
- Department of Medicine, Division of Endocrinology and Metabolism, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jaideep Behari
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Pittsburgh, PA 15261, USA
- UPMC Liver Clinic, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Alejandro Soto-Gutierrez
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Larry Vernetti
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Andrew Stern
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Albert Gough
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mark T Miedel
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - D Lansing Taylor
- University of Pittsburgh Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| |
Collapse
|
36
|
Chen P, Wu X, Gu X, Han J, Xue M, Liang X. FoxO1 in Micropterus salmoides: Molecular characterization and its roles in glucose metabolism by glucose or insulin-glucose loading. Gen Comp Endocrinol 2021; 310:113811. [PMID: 33979571 DOI: 10.1016/j.ygcen.2021.113811] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 04/15/2021] [Accepted: 05/07/2021] [Indexed: 12/18/2022]
Abstract
Forkhead box O1 (FoxO1), a nuclear transcription factor, plays an important role in insulin-mediated glucose metabolism. In this study, FoxO1 gene from largemouth bass (Micropterus salmoides) was cloned and characterized, and its effects on hepatic glucose metabolism regulated by insulin-AKT pathway were investigated in response to glucose or insulin-glucose injection. The full-length cDNA of FoxO1 consisted of 2541 bp and encoded 680 amino acids. Sequence alignments and phylogenetic analysis revealed that FoxO1 exhibited a high degree of conservation among teleost, retaining one forkhead domain, one transactivation domain, and three phosphorylation sites. FoxO1 mRNA was expressed in a wide range of tissues, and high in the brain and liver. Glucose loading resulted in persistent hyperglycemia, and plasma insulin levels remained unchanged except at 1 h. After the insulin-glucose injection, insulin levels were significantly elevated and glucose levels recovered to the basal value after 6 h, which indicated insufficient insulin secretion caused persistent hyperglycemia in this species. Compared with the glucose injection group, transcript levels and enzyme activities of hepatic glycolysis-related genes (GK and PK) were significantly activated, and gluconeogenesis-related genes (PEPCK and G6Pase) were significantly depressed at 3 h after the insulin-glucose injection. Besides, phosphorylation of AKT-FoxO1 pathway was significantly activated. Therefore, insulin improved glucose metabolism by activating the AKT-FoxO1 phosphorylation to decrease hyperglycemia stress after the meal, which indicated insufficient insulin secretion was the reason for glucose intolerance in largemouth bass. Meanwhile, conserved S267 and S329 phosphorylation sites of FoxO1 were confirmed to be regulated by AKT and mediated the glucose metabolism. In conclusion, activation of insulin-AKT-FoxO1 pathway improved glucose tolerance through mediating glucose metabolism in largemouth bass.
Collapse
Affiliation(s)
- Pei Chen
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xiufeng Wu
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xu Gu
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Juan Han
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing 100081, China
| | - Min Xue
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| | - Xiaofang Liang
- National Aquafeed Safety Assessment Center, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
37
|
Richter MM, Plomgaard P. The Regulation of Circulating Hepatokines by Fructose Ingestion in Humans. J Endocr Soc 2021; 5:bvab121. [PMID: 34337280 PMCID: PMC8317633 DOI: 10.1210/jendso/bvab121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Indexed: 01/22/2023] Open
Abstract
Context Fibroblast growth factor 21 (FGF21), follistatin, angiopoietin-like 4 (ANGPTL4), and growth differential factor 15 (GDF15) are regulated by energy metabolism. Recent findings in humans demonstrate that fructose ingestion increases circulating FGF21, with increased response in conditions of insulin resistance. Objective This study examines the acute effect of fructose and somatostatin on circulating FGF21, follistatin, ANGPTL4, and GDF15 in humans. Methods Plasma FGF21, follistatin, ANGPTL4, and GDF15 concentrations were measured in response to oral ingestion of 75 g of fructose in 10 young healthy males with and without a 15-minute infusion of somatostatin to block insulin secretion. A control infusion of somatostatin was also performed in the same subjects. Results Following fructose ingestion, plasma FGF21 peaked at 3.7-fold higher than basal concentration (P < 0.05), and it increased 4.9-fold compared with basal concentration (P < 0.05) when somatostatin was infused. Plasma follistatin increased 1.8-fold after fructose ingestion (P < 0.05), but this increase was blunted by concomitant somatostatin infusion. For plasma ANGPTL4 and GDF15, no increases were obtained following fructose ingestion. Infusion of somatostatin alone slightly increased plasma FGF21 and follistatin. Conclusion Here we show that in humans (1) the fructose-induced increase in plasma FGF21 was enhanced when somatostatin was infused, suggesting an inhibitory role of insulin on the fructose-induced FGF21 increase; (2) fructose ingestion also increased plasma follistatin, but somatostatin infusion blunted the increase; and (3) fructose ingestion had no stimulating effect on ANGPTL4 and GDF15 levels, demonstrating differences in the hepatokine response to fructose ingestion.
Collapse
Affiliation(s)
- Michael M Richter
- Department of Clinical Biochemistry, Rigshospitalet, DK-2100 Copenhagen, Denmark
| | - Peter Plomgaard
- Department of Clinical Biochemistry, Rigshospitalet, DK-2100 Copenhagen, Denmark.,The Centre of Inflammation and Metabolism and the Centre for Physical Activity Research, Department of Infectious Diseases and CMRC, Rigshospitalet, DK-2100 Copenhagen, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
38
|
Selig JI, Boulgaropoulos J, Niazy N, Ouwens DM, Preuß K, Horn P, Westenfeld R, Lichtenberg A, Akhyari P, Barth M. Crosstalk of Diabetic Conditions with Static Versus Dynamic Flow Environment-Impact on Aortic Valve Remodeling. Int J Mol Sci 2021; 22:ijms22136976. [PMID: 34203572 PMCID: PMC8268732 DOI: 10.3390/ijms22136976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/23/2021] [Indexed: 01/12/2023] Open
Abstract
Type 2 diabetes mellitus (T2D) is one of the prominent risk factors for the development and progression of calcific aortic valve disease. Nevertheless, little is known about molecular mechanisms of how T2D affects aortic valve (AV) remodeling. In this study, the influence of hyperinsulinemia and hyperglycemia on degenerative processes in valvular tissue is analyzed in intact AV exposed to an either static or dynamic 3D environment, respectively. The complex native dynamic environment of AV is simulated using a software-governed bioreactor system with controlled pulsatile flow. Dynamic cultivation resulted in significantly stronger fibrosis in AV tissue compared to static cultivation, while hyperinsulinemia and hyperglycemia had no impact on fibrosis. The expression of key differentiation markers and proteoglycans were altered by diabetic conditions in an environment-dependent manner. Furthermore, hyperinsulinemia and hyperglycemia affect insulin-signaling pathways. Western blot analysis showed increased phosphorylation level of protein kinase B (AKT) after acute insulin stimulation, which was lost in AV under hyperinsulinemia, indicating acquired insulin resistance of the AV tissue in response to elevated insulin levels. These data underline a complex interplay of diabetic conditions on one hand and biomechanical 3D environment on the other hand that possesses an impact on AV tissue remodeling.
Collapse
Affiliation(s)
- Jessica I. Selig
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - Joana Boulgaropoulos
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - Naima Niazy
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - D. Margriet Ouwens
- Institute of Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Auf’m Hennekamp 65, 40225 Düsseldorf, Germany;
- German Center for Diabetes Research (DZD), Ingolstädter Landstraße 1, Neuherberg, 85764 München, Germany
- Department of Endocrinology, Ghent University Hospital, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Karlheinz Preuß
- Faculty of Biotechnology, Bioprocessing, Modulation and Simulation, University of Applied Sciences Mannheim, Paul-Wittsack-Straße 10, 68163 Mannheim, Germany;
| | - Patrick Horn
- Department of Cardiology, Pneumology and Angiology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (P.H.); (R.W.)
| | - Ralf Westenfeld
- Department of Cardiology, Pneumology and Angiology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (P.H.); (R.W.)
| | - Artur Lichtenberg
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
- Correspondence:
| | - Mareike Barth
- Department of Cardiac Surgery, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225 Düsseldorf, Germany; (J.I.S.); (J.B.); (N.N.); (A.L.); (M.B.)
| |
Collapse
|
39
|
Guo X, Li X, Yang W, Liao W, Shen JZ, Ai W, Pan Q, Sun Y, Zhang K, Zhang R, Qiu Y, Dai Q, Zheng H, Guo S. Metformin Targets Foxo1 to Control Glucose Homeostasis. Biomolecules 2021; 11:biom11060873. [PMID: 34208360 PMCID: PMC8231152 DOI: 10.3390/biom11060873] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 11/28/2022] Open
Abstract
Metformin is the first-line pharmacotherapy for type 2 diabetes mellitus (T2D). Metformin exerts its glucose-lowering effect primarily through decreasing hepatic glucose production (HGP). However, the precise molecular mechanisms of metformin remain unclear due to supra-pharmacological concentration of metformin used in the study. Here, we investigated the role of Foxo1 in metformin action in control of glucose homeostasis and its mechanism via the transcription factor Foxo1 in mice, as well as the clinical relevance with co-treatment of aspirin. We showed that metformin inhibits HGP and blood glucose in a Foxo1-dependent manner. Furthermore, we identified that metformin suppresses glucagon-induced HGP through inhibiting the PKA→Foxo1 signaling pathway. In both cells and mice, Foxo1-S273D or A mutation abolished the suppressive effect of metformin on glucagon or fasting-induced HGP. We further showed that metformin attenuates PKA activity, decreases Foxo1-S273 phosphorylation, and improves glucose homeostasis in diet-induced obese mice. We also provided evidence that salicylate suppresses HGP and blood glucose through the PKA→Foxo1 signaling pathway, but it has no further additive improvement with metformin in control of glucose homeostasis. Our study demonstrates that metformin inhibits HGP through PKA-regulated transcription factor Foxo1 and its S273 phosphorylation.
Collapse
Affiliation(s)
- Xiaoqin Guo
- Xinqiao Hospital, Army Medical University, Chongqing 400037, China; (X.G.); (K.Z.); (R.Z.); (Y.Q.); (Q.D.)
| | - Xiaopeng Li
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
| | - Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
| | - Wang Liao
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
| | - James Zheng Shen
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
| | - Kebin Zhang
- Xinqiao Hospital, Army Medical University, Chongqing 400037, China; (X.G.); (K.Z.); (R.Z.); (Y.Q.); (Q.D.)
| | - Rui Zhang
- Xinqiao Hospital, Army Medical University, Chongqing 400037, China; (X.G.); (K.Z.); (R.Z.); (Y.Q.); (Q.D.)
| | - Yuyang Qiu
- Xinqiao Hospital, Army Medical University, Chongqing 400037, China; (X.G.); (K.Z.); (R.Z.); (Y.Q.); (Q.D.)
| | - Qian Dai
- Xinqiao Hospital, Army Medical University, Chongqing 400037, China; (X.G.); (K.Z.); (R.Z.); (Y.Q.); (Q.D.)
| | - Hongting Zheng
- Xinqiao Hospital, Army Medical University, Chongqing 400037, China; (X.G.); (K.Z.); (R.Z.); (Y.Q.); (Q.D.)
- Correspondence: (H.Z.); (S.G.)
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Science, Texas A&M University, College Station, TX 77843, USA; (X.L.); (W.Y.); (W.L.); (J.Z.S.); (W.A.); (Q.P.); (Y.S.)
- Correspondence: (H.Z.); (S.G.)
| |
Collapse
|
40
|
Zhang X, Jiang L, Liu H. Forkhead Box Protein O1: Functional Diversity and Post-Translational Modification, a New Therapeutic Target? DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:1851-1860. [PMID: 33976536 PMCID: PMC8106445 DOI: 10.2147/dddt.s305016] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 04/19/2021] [Indexed: 11/23/2022]
Abstract
Forkhead box protein O1 (FoXO1) is a transcription factor involved in the regulation of a wide variety of physiological process including glucose metabolism, lipogenesis, bone mass, apoptosis, and autophagy. FoXO1 dysfunction is involved in the pathophysiology of various diseases including metabolic diseases, atherosclerosis, and tumors. FoXO1 activity is regulated in response to different physiological or pathogenic conditions by changes in protein expression and post-translational modifications. Various modifications cooperate to regulate FoXO1 activity and FoXO1 target gene transcription. In this review, we summarize how different post-translational modifications regulate FoXO1 physiological function, which may provide new insights for drug design and development.
Collapse
Affiliation(s)
- Xiaojun Zhang
- Department of Cardiology, Shandong Rongjun General Hospital, Jinan, 250013, People's Republic of China
| | - Lusheng Jiang
- Department of Emergency, Shandong Rongjun General Hospital, Jinan, 250013, People's Republic of China
| | - Huimin Liu
- Blood Purification Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| |
Collapse
|
41
|
Ji L, Wang Q, Liu M, Zhu C, Xiao Y, Han J, Fang Y, Ye J, Yin J, Wei L. The 14-3-3 protein YWHAB inhibits glucagon-induced hepatic gluconeogenesis through interacting with the glucagon receptor and FOXO1. FEBS Lett 2021; 595:1275-1288. [PMID: 33641163 DOI: 10.1002/1873-3468.14063] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 01/15/2021] [Accepted: 01/23/2021] [Indexed: 01/09/2023]
Abstract
Glucagon antagonism has been reported as a new therapeutic approach to hyperglycaemia. As the 14-3-3 protein YWHAB has been identified as a regulator of the glucagon receptor (GCGR) by affinity purification and mass spectrometry, we examined the role of YWHAB in vivo. Ywhab knockout mice display impaired blood glucose homeostasis only under pyruvate stimulation. Deletion of Ywhab in mouse primary hepatocytes (MPHs) increases hepatocyte glucose production by magnifying the effect of glucagon. Mechanistic analysis indicates that YWHAB forms a phosphorylation-dependent complex with GCGR and directly interacts with forkhead box O1 (FOXO1). Together, these results reveal the inhibitory role of YWHAB in glucagon-mediated hepatic glucose production, which may be a potential target for the control of gluconeogenesis and associated metabolic diseases.
Collapse
Affiliation(s)
- Linlin Ji
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Qianqian Wang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
- Department of Endocrinology, School of Medicine, Shanghai Tongren Hospital, affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Mengdan Liu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Chaoyu Zhu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Yuanyuan Xiao
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Junfeng Han
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Yunyun Fang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Jianping Ye
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Jun Yin
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| | - Li Wei
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Center for Diabetes, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai, China
| |
Collapse
|
42
|
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
|
43
|
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: 0.5] [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
|
44
|
Vivar R, Anfossi R, Humeres C, Catalán M, Reyes C, Cárdenas S, Contreras A, Aránguiz P, González F, Diaz-Araya G. FoxO1 is required for high glucose-dependent cardiac fibroblasts into myofibroblast phenoconversion. Cell Signal 2021; 83:109978. [PMID: 33722671 DOI: 10.1016/j.cellsig.2021.109978] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022]
Abstract
In the normal heart, cardiac fibroblasts (CFs) maintain extracellular matrix (ECM) homeostasis, whereas in pathological conditions, such as diabetes mellitus (DM), CFs converse into cardiac myofibroblasts (CMFs) and this CFs phenoconversion increase the synthesis and secretion of ECM proteins, promoting cardiac fibrosis and heart dysfunction. High glucose (HG) conditions increase TGF-β1 expression and FoxO1 activity, whereas FoxO1 is crucial to CFs phenoconversion induced by TGF-β1. In addition, FoxO1 increases CTGF expression, whereas CTGF plays an active role in the fibrotic process induced by hyperglycemia. However, the role of FoxO1 and CTGF in CFs phenoconversion induced by HG is not clear. In this study, we investigated the effects of FoxO1 pharmacological inhibition on CFs phenoconversion in both in vitro and ex vivo models of DM. Our results demonstrate that HG induces CFs phenoconversion and FoxO1 activation. Moreover, AS1842856, a pharmacological inhibitor of FoxO1 activity, prevents CFs phenoconversion and CTGF expression increase induced by HG, whereas these results were corroborated by FoxO1 silencing. Additionally, K252a, a pharmacological blocker of CTGF receptor, prevents HG-induced CFs phenoconversion, which was corroborated with CTGF expression knockdown. Furthermore, through CFs isolation from heart of diabetic rats, we showed that hyperglycemia induces FoxO1 activation, the increase of CTGF expression and CFs phenoconversion, whereas the FoxO1 activity inhibition reverses the effects induced by hyperglycemia on CFs. Altogether, our results demonstrate that FoxO1 and CTGF are necessary for CFs phenoconversion induced by HG and suggest that both proteins are likely to become a potential targeted drug for fibrotic response induced by hyperglycemic conditions.
Collapse
Affiliation(s)
- Raúl Vivar
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Renatto Anfossi
- Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claudio Humeres
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Mabel Catalán
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Christopher Reyes
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Simone Cárdenas
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile.
| | - Alejandra Contreras
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pablo Aránguiz
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andrés Bello, 2520000 Viña del Mar, Chile
| | - Fabiola González
- Molecular and Clinical Pharmacology Program, Biomedical Science Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Guillermo Diaz-Araya
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.; Department of Pharmacological & Toxicological Chemistry, Faculty of Chemical & Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.
| |
Collapse
|
45
|
Liao W, Yang W, Shen Z, Ai W, Pan Q, Sun Y, Guo S. Heme Oxygenase-1 Regulates Ferrous Iron and Foxo1 in Control of Hepatic Gluconeogenesis. Diabetes 2021; 70:696-709. [PMID: 33408127 PMCID: PMC7897351 DOI: 10.2337/db20-0954] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
The liver is a key player for maintaining glucose homeostasis. Excessive hepatic glucose production is considered to be a key for the onset of type 2 diabetes. The primary function of heme oxygenase-1 (HO1) is to catalyze the degradation of heme into biliverdin, ferrous iron, and carbon monoxide. Previous studies have demonstrated that the degradation of heme by HO1 in the liver results in mitochondrial dysfunction and drives insulin resistance. In this study, by overexpressing HO1 in hepatocytes and mice, we showed that HO1 promotes gluconeogenesis in a Foxo1-dependent manner. Importantly, HO1 overexpression increased the generation of ferrous iron in the liver, which further activates nuclear factor-κB and phosphorylates Foxo1 at Ser273 to enhance gluconeogenesis. We further assessed the role of HO1 in insulin-resistant liver-specific knockout of IRS1 and IRS2 genes (L-DKO) mice, which exhibit upregulation of HO1 in the liver and hepatic ferrous iron overload. HO1 knockdown by shRNA or treatment of iron chelator rescued the aberrant gluconeogenesis in L-DKO mice. In addition, we found that systemic iron overload promotes gluconeogenesis by activating the hepatic protein kinase A→Foxo1 axis. Thus, our results demonstrate the role of HO1 in regulating hepatic iron status and Foxo1 to control gluconeogenesis and blood glucose.
Collapse
Affiliation(s)
- Wang Liao
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Zheng Shen
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| |
Collapse
|
46
|
Alterations of Gut Microbiota by Overnutrition Impact Gluconeogenic Gene Expression and Insulin Signaling. Int J Mol Sci 2021; 22:ijms22042121. [PMID: 33672754 PMCID: PMC7924631 DOI: 10.3390/ijms22042121] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 02/04/2023] Open
Abstract
A high-fat, Western-style diet is an important predisposing factor for the onset of type 2 diabetes and obesity. It causes changes in gut microbial profile, reduction of microbial diversity, and the impairment of the intestinal barrier, leading to increased serum lipopolysaccharide (endotoxin) levels. Elevated lipopolysaccharide (LPS) induces acetyltransferase P300 both in the nucleus and cytoplasm of liver hepatocytes through the activation of the IRE1-XBP1 pathway in the endoplasmic reticulum stress. In the nucleus, induced P300 acetylates CRTC2 to increase CRTC2 abundance and drives Foxo1 gene expression, resulting in increased expression of the rate-limiting gluconeogenic gene G6pc and Pck1 and abnormal liver glucose production. Furthermore, abnormal cytoplasm-appearing P300 acetylates IRS1 and IRS2 to disrupt insulin signaling, leading to the prevention of nuclear exclusion and degradation of FOXO1 proteins to further exacerbate the expression of G6pc and Pck1 genes and liver glucose production. Inhibition of P300 acetyltransferase activity by chemical inhibitors improved insulin signaling and alleviated hyperglycemia in obese mice. Thus, P300 acetyltransferase activity appears to be a therapeutic target for the treatment of type 2 diabetes and obesity.
Collapse
|
47
|
Kim H, Lee DS, An TH, Park TJ, Lee EW, Han BS, Kim WK, Lee CH, Lee SC, Oh KJ, Bae KH. GADD45β Regulates Hepatic Gluconeogenesis via Modulating the Protein Stability of FoxO1. Biomedicines 2021; 9:biomedicines9010050. [PMID: 33435535 PMCID: PMC7827134 DOI: 10.3390/biomedicines9010050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/26/2020] [Accepted: 01/07/2021] [Indexed: 01/07/2023] Open
Abstract
Increased hepatic gluconeogenesis is one of the main contributors to the development of type 2 diabetes. Recently, it has been reported that growth arrest and DNA damage-inducible 45 beta (GADD45β) is induced under both fasting and high-fat diet (HFD) conditions that stimulate hepatic gluconeogenesis. Here, this study aimed to establish the molecular mechanisms underlying the novel role of GADD45β in hepatic gluconeogenesis. Both whole-body knockout (KO) mice and adenovirus-mediated knockdown (KD) mice of GADD45β exhibited decreased hepatic gluconeogenic gene expression concomitant with reduced blood glucose levels under fasting and HFD conditions, but showed a more pronounced effect in GADD45β KD mice. Further, in primary hepatocytes, GADD45β KD reduced glucose output, whereas GADD45β overexpression increased it. Mechanistically, GADD45β did not affect Akt-mediated forkhead box protein O1 (FoxO1) phosphorylation and forskolin-induced cAMP response element-binding protein (CREB) phosphorylation. Rather it increased FoxO1 transcriptional activity via enhanced protein stability of FoxO1. Further, GADD45β colocalized and physically interacted with FoxO1. Additionally, GADD45β deficiency potentiated insulin-mediated suppression of hepatic gluconeogenic genes, and it were impeded by the restoration of GADD45β expression. Our finding demonstrates GADD45β as a novel and essential regulator of hepatic gluconeogenesis. It will provide a deeper understanding of the FoxO1-mediated gluconeogenesis.
Collapse
Affiliation(s)
- Hyunmi Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Da Som Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Tae Hyeon An
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Tae-Jun Park
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
| | - Baek Soo Han
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Chul-Ho Lee
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea;
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
- Correspondence: (K.-J.O.); (K.-H.B.)
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea; (H.K.); (D.S.L.); (T.H.A.); (T.-J.P.); (E.-W.L.); (B.S.H.); (W.K.K.); (S.C.L.)
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141, Korea
- Correspondence: (K.-J.O.); (K.-H.B.)
| |
Collapse
|
48
|
He Q, Luo J, Wu J, Yao W, Li Z, Wang H, Xu H. FoxO1 Knockdown Promotes Fatty Acid Synthesis via Modulating SREBP1 Activities in the Dairy Goat Mammary Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12067-12078. [PMID: 33054209 DOI: 10.1021/acs.jafc.0c05237] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
FoxO1 is a crucial transcription factor involved in lipid metabolism in mouse liver through repressing a key regulator of lipogenesis, sterol regulatory element binding protein 1 (SREBP1). However, it remains elusive whether FoxO1 plays roles in the regulation of fatty acid metabolism during lactation in dairy goats. In this study, we aim to investigate the function of FoxO1 in goat mammary epithelial cells (GMECs). We found that the expression of FoxO1 is significantly upregulated during lactation compared with the dry period. FoxO1 knockdown enhanced the expression of genes related to de novo fatty acid synthesis (e.g., FASN, ELOVL6 and SCD1) and triacylglycerol (TAG) synthesis (e.g., DGAT2 and GPAM). Consistently, intracellular TAG was significantly increased in FoxO1 knockdown cells and reduced in FoxO1 overexpression cells. Immunofluorescence staining revealed that insulin suppresses FoxO1 transcription by promoting its nuclear export. Further, we found that FoxO1 inhibits insulin-induced SREBP1 promoter activities in GMECs. Moreover, FoxO1 suppresses SREBP1 transcription via the LXR response element (LXRE) and SREBP response element (SRE) located in the SREBP1 promoter. Our data reveal that FoxO1 plays critical roles in regulating the synthesis of the fatty acid and triacylglycerol (TAG) in GMECs.
Collapse
Affiliation(s)
- Qiuya He
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jun Luo
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Jiao Wu
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Weiwei Yao
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Zhuang Li
- Shaanxi Key Laboratory of Molecular Biology for Agriculture, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Hui Wang
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu, Sichuan 610041, PR China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
49
|
Selective inhibition of CBP/p300 HAT by A-485 results in suppression of lipogenesis and hepatic gluconeogenesis. Cell Death Dis 2020; 11:745. [PMID: 32917859 PMCID: PMC7486386 DOI: 10.1038/s41419-020-02960-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 08/09/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
Abstract
The histone acetyltransferases CREB-binding protein (CBP) and its paralogue p300 are transcriptional coactivators which are essential for a multitude of signaling pathways and energy homeostasis. However, the role of CBP/p300 HAT domain in regulating energy balance is still unclear. Here, C57BL/6 mice fed with either normal chow diet (NCD) or high-fat diet (HFD) were administrated with A-485, a recently reported selective inhibitor of CBP/p300 HAT activity for 1 week and the metabolic change was analyzed. The white adipose tissue (WAT) weight and adipocyte size were reduced in A-485-administrated mice, with decreased expressions of lipogenic genes and transcriptional factors. In the liver of A-485-treated mice, the lipid content and lipogenic gene expressions were lowered while the binding of forkhead box O1 (FOXO1) to glucose-6-phosphatase (G6Pc) promoter was reduced, leading to decreased expression of G6Pc. In primary mouse hepatocytes, A-485 abolished cAMP-elicited mRNA expressions of key gluconeogenic enzymes and promoted FOXO1 protein degradation via increasing its ubiquitination. Thus, A-485 inhibits lipogenesis in WAT and liver as well as decreases hepatic glucose production via preventing FOXO1 acetylation, leading to its protein degradation through a proteasome-dependent pathway. The specific inhibition of CBP/p300 HAT will provide a novel therapeutic approach for metabolic diseases.
Collapse
|
50
|
Abstract
Forkhead box O (FOXO) transcription factors regulate diverse biological processes, affecting development, metabolism, stem cell maintenance and longevity. They have also been increasingly recognised as tumour suppressors through their ability to regulate genes essential for cell proliferation, cell death, senescence, angiogenesis, cell migration and metastasis. Mechanistically, FOXO proteins serve as key connection points to allow diverse proliferative, nutrient and stress signals to converge and integrate with distinct gene networks to control cell fate, metabolism and cancer development. In consequence, deregulation of FOXO expression and function can promote genetic disorders, metabolic diseases, deregulated ageing and cancer. Metastasis is the process by which cancer cells spread from the primary tumour often via the bloodstream or the lymphatic system and is the major cause of cancer death. The regulation and deregulation of FOXO transcription factors occur predominantly at the post-transcriptional and post-translational levels mediated by regulatory non-coding RNAs, their interactions with other protein partners and co-factors and a combination of post-translational modifications (PTMs), including phosphorylation, acetylation, methylation and ubiquitination. This review discusses the role and regulation of FOXO proteins in tumour initiation and progression, with a particular emphasis on cancer metastasis. An understanding of how signalling networks integrate with the FOXO transcription factors to modulate their developmental, metabolic and tumour-suppressive functions in normal tissues and in cancer will offer a new perspective on tumorigenesis and metastasis, and open up therapeutic opportunities for malignant diseases.
Collapse
Affiliation(s)
- Yannasittha Jiramongkol
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK.
| |
Collapse
|