1
|
Walls KM, Joh JY, Martinez MM, Hong KU, Hein DW. Metabolic effects of heterocyclic amines on insulin‑induced AKT phosphorylation and gluconeogenic gene expression are modified by N -acetyltransferase 2 genetic polymorphism. Pharmacogenet Genomics 2025; 35:119-126. [PMID: 39878101 PMCID: PMC12043411 DOI: 10.1097/fpc.0000000000000559] [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] [Indexed: 01/31/2025]
Abstract
OBJECTIVE Heterocyclic amines (HCAs) are mutagens and carcinogens primarily generated when cooking meat at high temperatures or until well-done, and their major metabolic pathway includes hepatic N -hydroxylation via CYP1A2 followed by O -acetylation via N -acetyltransferase 2 (NAT2). NAT2 expresses a well-defined genetic polymorphism in humans resulting in rapid and slow acetylators. Recent epidemiological studies reported significant associations between dietary HCA exposure and insulin resistance and type II diabetes. METHODS We assessed the effect of some of the most common HCAs found in cooked meat, 2-amino-3,4-dimethylimidazo[4,5-f]quinoline, 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline, and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine, on insulin signaling and gluconeogenic gene expression in cryopreserved human hepatocytes characterized by their NAT2 genotype and phenotype to investigate the role of NAT2 genetic polymorphism in HCA-induced metabolic dysregulation. RESULTS HCA treatment significantly reduced insulin-induced protein kinase B phosphorylation and significantly increased expression of genes involved in gluconeogenesis ( G6PC , PCK1 , FOXO1 , and PPARA ) in cryopreserved human hepatocytes from rapid but not from slow acetylators. CONCLUSION The findings suggest that NAT2 genetic polymorphism modifies HCA-induced insulin resistance and gluconeogenic gene expression, implying that individuals with rapid acetylator phenotype may be at greater risk of dysregulated glucose homeostasis following exposure to HCAs.
Collapse
Affiliation(s)
- Kennedy M Walls
- Department of Pharmacology & Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | | | | | | | | |
Collapse
|
2
|
Choudhuri S, Garg NJ. Hepatocyte Nuclear Factor 4 Alpha: A Key Regulator of Liver Disease Pathology and Haemostatic Disorders. Liver Int 2025; 45:e16245. [PMID: 40387433 DOI: 10.1111/liv.16245] [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: 08/15/2024] [Revised: 12/19/2024] [Accepted: 01/03/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE Hepatocyte nuclear factor 4 alpha (HNF4α) is a master regulator of hepatocyte differentiation in fetal and adult liver and exerts its transcriptional role in determining physiological functions of the liver. The objective of this review is to address the current knowledge of molecular mechanisms involved in HNF4α regulation in multiple aspects of liver disease pathogenesis. METHODS Based on available literature, this review summarises the current state of knowledge onthe mechanism of HNF4α dysregulation, and the role of HNF4α activity inregulating early to advanced stages of various liver diseases. RESULTS Patients with deranged HNF4α expression are at higher risk for the development of liver diseases such as viral hepatitis, alcoholic/nonalcoholic fatty liver disease, hepatocellular carcinoma, and haematological disorders such as coagulopathy and bleeding disorders. DISCUSSION HNF4α interactions with nuclear receptors and target genes promote liver disease pathology by regulating various metabolic pathways. The strong correlation between deranged HNF4α expression and the severity of liver diseases suggests that targeting HNF4α expression can offer potential therapeutic strategy in the prevention of liver disease pathology and haemostatic disorders.
Collapse
Affiliation(s)
- Subhadip Choudhuri
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
- Institute for Human Infections and Immunity (IHII), University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| | - Nisha Jain Garg
- Department of Microbiology and Immunology, University of Texas Medical Branch (UTMB), Galveston, Texas, USA
- Institute for Human Infections and Immunity (IHII), University of Texas Medical Branch (UTMB), Galveston, Texas, USA
| |
Collapse
|
3
|
Yenmis G, Kallenci I, Dokur M, Koc S, Yalinkilic SB, Atak E, Demirbilek M, Arkan H. The Distribution of Sport Performance Gene Variations Through COVID-19 Disease Severity. Diagnostics (Basel) 2025; 15:701. [PMID: 40150043 PMCID: PMC11941099 DOI: 10.3390/diagnostics15060701] [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: 01/05/2025] [Revised: 02/23/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Since its emergence in 2020, researchers worldwide have been collaborating to better understand the SARS-CoV-2 disease's pathophysiology. Disease severity can vary based on several factors, including comorbidities and genetic variations. Notably, recent studies have highlighted the role of genes associated with athletic performance, such as ACE, ACTN3, and PPARGC1A, in influencing muscle function, cardiovascular health, and the body's metabolic response. Given that these genes also impact oxidative metabolism, inflammation, and respiratory efficiency, we hypothesized that they might play a critical role in the host's response to SARS-CoV-2 infection. This study aimed to investigate the association between disease severity and genetic polymorphisms in these sport performance-related genes, specifically ACE rs4646994, ACTN3 rs1815739, and PPARGC1A rs8192678. Methods: A total of 422 COVID-19-positive patients were included in this study. The participants were divided into three groups: a severe group (77 patients) requiring intensive care unit (ICU) admission, a mild group (300 patients) exhibiting at least one symptom, and an asymptomatic control group. Genotyping was performed using restriction fragment length polymorphism PCR. Results: The D allele and DD genotype of ACE and the T allele and TT genotype of ACTN3 were found to confer protective effects against severe SARS-CoV-2 infection. Conversely, the PPARGC1A TC genotype and the ACE-PPARGC1A ins/ins + TC combined genotype were associated with increased disease severity (p < 0.05). Conclusions: Although vaccination has reduced the severity of SARS-CoV-2, the virus continues to impact human health. Inter-individual differences due to these genetic variations will broaden the horizon of knowledge on the pathophysiology of the disease.
Collapse
Affiliation(s)
- Guven Yenmis
- Department of Medical Biology, Tayfur Ata Sokmen School of Medicine, Hatay Mustafa Kemal University, Hatay 31060, Turkey
| | - Ilayda Kallenci
- Department of Molecular Biology and Genetics, Faculty of Natural Sciences and Engineering, Biruni University, Istanbul 34015, Turkey; (I.K.); (S.B.Y.)
| | - Mehmet Dokur
- Department of Emergency Medicine, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik 11230, Turkey;
| | - Suna Koc
- Department of Anesthesia and Reanimation, School of Medicine, Biruni University, Istanbul 34015, Turkey;
| | - Sila Basak Yalinkilic
- Department of Molecular Biology and Genetics, Faculty of Natural Sciences and Engineering, Biruni University, Istanbul 34015, Turkey; (I.K.); (S.B.Y.)
| | - Evren Atak
- Department of Bioinformatics and System Biology, Institute of Natural and Applied Sciences, Gebze Technical University, Kocaeli 41400, Turkey;
| | - Mahmut Demirbilek
- Department of Emergency Medicine, School of Medicine, Biruni University, Istanbul 34015, Turkey;
| | - Hulya Arkan
- Department of Biotechnology, Institute of Science, Yildiz Technical University, Istanbul 34210, Turkey;
| |
Collapse
|
4
|
Kubota N, Kubota T, Kadowaki T. Physiological and pathophysiological actions of insulin in the liver. Endocr J 2025; 72:149-159. [PMID: 39231651 PMCID: PMC11850106 DOI: 10.1507/endocrj.ej24-0192] [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] [Received: 04/08/2024] [Accepted: 06/21/2024] [Indexed: 09/06/2024] Open
Abstract
The liver plays an important role in the control of glucose homeostasis. When insulin levels are low, such as in the fasting state, gluconeogenesis and glycogenolysis are stimulated to maintain the blood glucose levels. Conversely, in the presence of increased insulin levels, such as after a meal, synthesis of glycogen and lipid occurs to maintain the blood glucose levels within normal range. Insulin receptor signaling regulates glycogenesis, gluconeogenesis and lipogenesis through downstream pathways such as the insulin receptor substrate (IRS)-phosphoinositide 3 (PI3) kinase-Akt pathway. IRS-1 and IRS-2 are abundantly expressed in the liver and are thought to be responsible for transmitting the insulin signal from the insulin receptor to the intracellular effectors involved in the regulation of glucose and lipid homeostasis. Impaired insulin receptor signaling can cause hepatic insulin resistance and lead to type 2 diabetes. In the present study, we focus on a concept called "selective insulin resistance," which has received increasing attention recently: the frequent coexistence of hyperglycemia and hepatic steatosis in people with type 2 diabetes and obesity suggests that it is possible for the insulin signaling regulating gluconeogenesis to be impaired even while that regulating lipogenesis is preserved, suggestive of selective insulin resistance. In this review, we review the progress in research on the insulin actions and insulin signaling in the liver.
Collapse
Affiliation(s)
- Naoto Kubota
- Department of Metabolic Medicine, Faculty of Life Sciences, Kumamoto University, Kumamoto 860-8556, Japan
| | - Tetsuya Kubota
- Division of Diabetes and Metabolism, The Institute of Medical Science, Asahi Life Foundation, Tokyo 103-0002, Japan
| | | |
Collapse
|
5
|
Sartorelli V, Ciuffoli V. Metabolic regulation in adult and aging skeletal muscle stem cells. Genes Dev 2025; 39:186-208. [PMID: 39662967 PMCID: PMC11789647 DOI: 10.1101/gad.352277.124] [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] [Indexed: 12/13/2024]
Abstract
Adult stem cells maintain homeostasis and enable regeneration of most tissues. Quiescence, proliferation, and differentiation of stem cells and their progenitors are tightly regulated processes governed by dynamic transcriptional, epigenetic, and metabolic programs. Previously thought to merely reflect a cell's energy state, metabolism is now recognized for its critical regulatory functions, controlling not only energy and biomass production but also the cell's transcriptome and epigenome. In this review, we explore how metabolic pathways, metabolites, and transcriptional and epigenetic regulators are functionally interlinked in adult and aging skeletal muscle stem cells.
Collapse
Affiliation(s)
- Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Veronica Ciuffoli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
6
|
Zhao J, Zhong Y, Huang Q, Pan Z, Zheng Y, Miao D, Liu S, Chen P, Liu C, Liu M, Shen C. Cassia mimosoides L. decoction improves non-alcoholic fatty liver disease by modulating the pregnane X receptor. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119199. [PMID: 39631715 DOI: 10.1016/j.jep.2024.119199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/29/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cassia mimosoides L. (CML) is a traditional Chinese medicine (TCM), which is frequently used in the clinical practice of TCM in the Lingnan region of China for the treatment of obesity. However, it is not clear whether decoction of cassia seeds has beneficial effects on non-alcoholic fatty liver disease (NAFLD). OBJECTIVES This study investigates the effect of CML on NAFLD and its underlying mechanisms. MATERIALS AND METHODS The high-fat diet (HFD) was used to induce NAFLD mice, and 40 male C57BL/6J mice were divided into Control, HFD, and CML groups (CML-low 1.5 g/kg, CML-medium 2.25 g/kg, CML-high 4.5 g/kg). The mouse primary hepatocytes (MPHs) of wild type (WT) and PXR-/- mice were induced using OAPA and divided into Control, OAPA, and CML groups (10 mg/L, 100 mg/L). Glycolipid metabolism, inflammation, and oxidative stress levels were detected in vivo and in vitro. RESULTS Compared to the HFD group, the CML groups demonstrated reduced body weight, triglycerides, total cholesterol, blood glucose, and mRNA levels of the lipid metabolism genes Srebp-1c and ACC1 in mice (p < 0.05 or 0.01). The ELISA results indicated that CML inhibited the production of IL-1β, IL-6, and TNF-α (p < 0.05). Furthermore, CML increased the SOD level (p < 0.01) to improve oxidative stress. RNA-seq expression showed that CML suppressed the transcriptional level of pregnane X receptor (PXR)(p < 0.05). In vitro experiments, the protective effect of CML against OAPA-induced lipid accumulation and inflammation observed in WT MPHs disappeared in PXR-/- MPHs (IC50: 1.04 mg/mL). CONCLUSION CML decoction ameliorates NAFLD mainly by inhibiting the PXR signaling pathway, which provides a theoretical basis for the broad application of CML in clinical practice.
Collapse
Affiliation(s)
- Jian Zhao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanhua Zhong
- Department of Acupuncture-Rehabilitation, Guangzhou-Liwan Hospital of Chinese Medicine, Guangzhou, China
| | - Qingyin Huang
- Department of Endocrinology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhisen Pan
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yi Zheng
- The Second Affiliated Hospital of Zhejiang Chinese Medical University, Xinhua Hospital of Zhejiang Province, Hangzhou, China
| | - Deyu Miao
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Siqi Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Penglong Chen
- Pharmacy Department of the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Changhui Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Min Liu
- Department of Endocrinology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuangpeng Shen
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China; Department of Endocrinology, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China; ShenShan Hospital, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Shanwei, 516600, China; Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
7
|
Zhang T, Zhao S, Gu C. Role of PGC-1α in the proliferation and metastasis of malignant tumors. J Mol Histol 2025; 56:77. [PMID: 39881043 DOI: 10.1007/s10735-025-10360-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 01/19/2025] [Indexed: 01/31/2025]
Abstract
Malignant tumors are among the major diseases threatening human survival in the world, and advancements in medical technology have led to a steady increase in their detection rates worldwide. Despite unique clinical presentations across the spectrum of malignancies, treatment modalities generally adhere to common strategies, encompassing primarily surgical intervention, radiation therapy, chemotherapy, and targeted treatments. Uncovering the genetic elements contributing to cancer cell proliferation, metastasis, and drug resistance remains a pivotal pursuit in the development of novel targeted therapeutics. Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A/PGC-1α) is a transcriptional coactivator that influences most cellular metabolic pathways. Its aberrant expression is associated with numerous chronic diseases, including diabetes, heart failure, neurodegenerative disorders, and cancer development. This study primarily discusses the structure, physiological functions, regulatory mechanisms, and research advancement concerning the role of PGC-1α in the proliferation and metastasis of malignant tumors. Targeting PGC-1α and its related regulatory pathways for therapeutic interventions holds promise in facilitating precise and individualized oncological treatments. This approach is expected to counteract drug resistance in patients with cancer and offer a novel direction for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Tianyi Zhang
- Department of Thoracic Surgery, Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
- Dalian Medical University, Dalian, 116011, China
| | - Shilei Zhao
- Department of Thoracic Surgery, Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
- Dalian Medical University, Dalian, 116011, China
| | - Chundong Gu
- Department of Thoracic Surgery, Lung Cancer Diagnosis and Treatment Center of Dalian, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
- Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
8
|
Chen J, Wu Y, Hao W, You J, Wu L. Non-canonical hepatic androgen receptor mediates glucagon sensitivity in female mice through the PGC1α/ERRα/mitochondria axis. Cell Rep 2025; 44:115188. [PMID: 39792556 DOI: 10.1016/j.celrep.2024.115188] [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: 07/19/2024] [Revised: 10/27/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Glucagon has recently been found to modulate liver fat content, in addition to its role in regulating gluconeogenesis. However, the precise mechanisms by which glucagon signaling synchronizes glucose and lipid metabolism in the liver remain poorly understood. By employing chemical and genetic approaches, we demonstrate that inhibiting the androgen receptor (AR) impairs the ability of glucagon to stimulate gluconeogenesis and lipid catabolism in primary hepatocytes and female mice. Notably, AR expression in the liver of female mice is up to three times higher than that in their male littermates, accounting for the more pronounced response to glucagon in females. Mechanistically, hepatic AR promotes energy metabolism and enhances lipid breakdown for liver glucose production in response to glucagon treatment through the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α)/estrogen-related receptor alpha (ERRα)-mitochondria axis. Overall, our findings highlight the crucial role of hepatic AR in mediating glucagon signaling and the sexual dimorphism in hepatic glucagon sensitivity.
Collapse
Affiliation(s)
- Jie Chen
- Fudan University, Shanghai, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Yuanyuan Wu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Wenyuan Secondary School Affiliated to Xuejun High School, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Pharmaceuticals, Hangzhou, Zhejiang, China
| | - Jia You
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| | - Lianfeng Wu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Luo X, Liu F, Zhu L, Liu C, Shen R, Ding X, Wang Y, Tang X, Peng Y, Zhang Z. Leupaxin promotes hepatic gluconeogenesis and glucose metabolism by coactivation with hepatic nuclear factor 4α. Mol Metab 2025; 91:102075. [PMID: 39603504 PMCID: PMC11647654 DOI: 10.1016/j.molmet.2024.102075] [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] [Received: 09/02/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND As the primary source of glucose during fasting, hepatic gluconeogenesis is rigorously regulated to maintain euglycemia. Abnormal gluconeogenesis in the liver can lead to hyperglycemia, a key diagnostic marker and the primary pathological contributor to type 2 diabetes (T2D) and metabolic disorders. Hepatic nuclear factor-4 (HNF4α) is an important regulator of gluconeogenesis. In this study, we identify leupaxin (LPXN) as a novel coactivator for HNF4α. Although previous studies have shown that LPXN is highly correlated with cancer types such as B-cell differentiation and hepatocellular carcinoma progression, the role of LPXN in gluconeogenesis remains unknown. METHODS We initially used protein pull-down assays, mass spectrometry and luciferase assays to identify the coactivator that interacts with HNF4α in gluconeogenesis. We further leveraged cell cultures and mouse models to validate the functional importance of molecular pathway during gluconeogenesis by using adenovirus-mediated overexpression and adeno-associated virus shRNA-mediated knockdown both in vivo and ex vivo, such as in ob/db/DIO mice, HepG2 and primary hepatocytes. Following, we used CUT&Tag and chip qPCR to identify the LPXN-mediated mechanisms underlying the observed abnormal gluconeogenesis. Additionally, we assessed the translational relevance of our findings using human liver tissues from both healthy donors and patients with obesity/type 2 diabetes. RESULTS We found that LPXN interacts with HNF4α to participate in gluconeogenesis. Knockdown of LPXN expression in the liver effectively enhanced glucose metabolism, while its overexpression in the liver effectively inhibited it. Mechanistically, LPXN could translocate into the nucleus and was essential for regulating gluconeogenesis by binding to the PEPCK promoter, which controlled the expression of an enzyme involved in gluconeogenesis, mainly through the Gcg-cAMP-PKA pathway. Additionally, LPXN expression was found to be increased in the livers of patients with steatosis and diabetes, supporting a pathological role of LPXN. CONCLUSIONS Taken together, our study provides evidence that LPXN plays a critical role in modulating hepatic gluconeogenesis, thereby reinforcing the fact that targeting LPXN may be a potential approach for the treatment of diabetes and metabolic disorders.
Collapse
Affiliation(s)
- Xiaomin Luo
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fang Liu
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Lijun Zhu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China; Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caizhi Liu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Ruhui Shen
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyin Ding
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yufan Wang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaofang Tang
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yongde Peng
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhijian Zhang
- Department of Endocrinology and Metabolism, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Deguchi K, Ushiroda C, Kamei Y, Kondo K, Tsuchida H, Seino Y, Yabe D, Suzuki A, Nagao S, Iizuka K. Glucose and Insulin Differently Regulate Gluconeogenic and Ureagenic Gene Expression. J Nutr Sci Vitaminol (Tokyo) 2025; 71:46-54. [PMID: 40024748 DOI: 10.3177/jnsv.71.46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Glucose and insulin positively regulate glycolysis and lipogenesis through the activation of carbohydrate response element-binding protein (ChREBP) and sterol regulatory element-binding protein 1c (SREBP1c), but their respective roles in the regulation of gluconeogenic and ureagenic genes remain unclear. We compared the effects of the insulin antagonist S961 and Chrebp deletion on hepatic glycolytic, lipogenic, gluconeogenic, and ureagenic gene expression in mice. S961 markedly increased the plasma glucose, insulin, and 3-OH-butyrate concentrations and reduced the hepatic triglyceride content, but Chrebp deletion had no additive effect. We subsequently estimated the expression of genes involved in the pathways of glycolysis, gluconeogenesis, and lipogenesis. S961 potently decreased both Chrebp and Srebf1c, but Chrebp deletion weakly decreased Srebf1c mRNA expression. Both the S961 and Chrebp deletion caused decreases in glycolytic (Gck and Pklr) and lipogenic (Fasn, Scd1, Me1, Spot14, Elovl6) gene expression. S961 increased the expression of many gluconeogenic genes (G6pc, Fbp1, Aldob, Slc37a4, Pck), whereas Chrebp deletion reduced the expression of gluconeogenic genes other than Pck1. Finally, we checked the metabolites and gene expression in the ureagenesis pathway. S961 increased ureagenic gene (Arg1, Asl, Ass1, Cps1, Otc) expression, which was consistent with the metabolite data: there were reductions in the concentrations of glutamate and aspartate and increases in those of citrulline, ornithine, urea, and proline. However, Chrebp deletion had no additive effect on ureagenesis. In conclusion, insulin rather than glucose regulate ureagenic gene expression, whereas glucose and insulin regulate gluconegenic gene expression in opposite directions.
Collapse
Affiliation(s)
- Kanako Deguchi
- Department of Clinical Nutrition, Fujita Health University
| | | | - Yuka Kamei
- Advanced Medical Research Center for Animal Models of Human Diseases, Fujita Health University
| | | | - Hiromi Tsuchida
- Department of Diabetes, Endocrinology and Metabolism, Gifu University Graduate School of Medicine
| | - Yusuke Seino
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism, Gifu University Graduate School of Medicine
- Center for One Medicine Innovative Translational Research, Gifu University
| | - Atsushi Suzuki
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University
| | - Shizuko Nagao
- Advanced Medical Research Center for Animal Models of Human Diseases, Fujita Health University
| | - Katsumi Iizuka
- Department of Clinical Nutrition, Fujita Health University
- Food and Nutrition Service Department, Fujita Health University Hospital
| |
Collapse
|
11
|
Okuno Y, Fukuhara A, Shimomura I. The role of oxidative stress, glucocorticoid receptor and ARMC5 in lipid metabolism. Endocr J 2024; 71:1097-1101. [PMID: 38925988 PMCID: PMC11778357 DOI: 10.1507/endocrj.ej24-0177] [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] [Received: 03/25/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Lipid metabolism includes lipogenesis, lipolysis, and cholesterol metabolism and it exerts a wide range of biological effects. We previously found novel roles of adipocyte oxidative stress in diet-induced obesity, adipocyte glucocorticoid receptor in Cushing syndrome, and ARMC5 in adrenocortical cells. Using genetically modified mice in which oxidative stress was eliminated or augmented specifically in adipose tissues, we have been able to elucidate that obesity-induced oxidative stress inhibited healthy adipose expansion and ameliorated insulin sensitivity. Using adipocyte-specific glucocorticoid receptor knockout mice, we found that glucocorticoids also inhibited healthy adipose expansion and decreased insulin sensitivity. This was partly due to the transcriptional upregulation of ATGL. We identified ARMC5 as a novel ubiquitin E3 ligase of full-length SREBF, a master regulator of lipid metabolism. In adrenocortical cells, ARMC5 suppresses SREBF2 activity, and loss of ARMC5 may lead to cholesterol accumulation and the development of primary bilateral macronodular adrenal hyperplasia.
Collapse
Affiliation(s)
- Yosuke Okuno
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Atsunori Fukuhara
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
- Department of Adipose Management, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Osaka University Graduate School of Medicine, Osaka 565-0871, Japan
| |
Collapse
|
12
|
Bai Y, Wen H, Lin J, Liu X, Yu H, Wu M, Wang L, Chen D. Tanshinone I improves renal fibrosis by promoting gluconeogenesis through upregulation of peroxisome proliferator-activated receptor-γ coactivator 1α. Ren Fail 2024; 46:2433710. [PMID: 39648664 PMCID: PMC11632924 DOI: 10.1080/0886022x.2024.2433710] [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: 09/04/2024] [Revised: 10/12/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024] Open
Abstract
BACKGROUND Renal fibrosis, a hallmark of chronic kidney disease, is closely associated with dysregulated gluconeogenesis. Tanshinone I (Tan I), a bioactive compound derived from the traditional Chinese medicine Danshen, exhibits antifibrotic and anti-inflammatory properties. However, its effects on gluconeogenesis and the mechanisms through which it alleviates renal fibrosis remain unclear. This study aimed to investigate whether Tan I promotes gluconeogenesis and mitigates renal fibrosis. METHODS Both in vivo and in vitro experiments were conducted. A unilateral ureteral obstruction (UUO) mouse model was used. Masson's trichrome, HE, and immunofluorescence staining, along with Western blotting, were employed. Lactate concentrations and a pyruvate tolerance test were conducted to assess glucose metabolism. In vitro, HK2 cells and primary renal tubular cells were treated with transforming growth factor-β (TGFβ) to induce fibrosis, and the effects of Tan I on glucose and lactate levels were examined. RESULTS In the UUO model, Tan I reduced fibrosis, decreased lactate accumulation, and modulated fibrosis markers while upregulating gluconeogenesis markers. Tanshinone I restored impaired renal gluconeogenesis, as evidenced by increased pyruvate levels. In vitro, Tan I inhibited fibrosis, reduced lactate levels, and increased glucose levels in cell supernatants. It also restored gluconeogenesis protein expression and decreased fibrotic protein levels. Peroxisome proliferator-activated receptor-γ coactivator (PGC1α) expression was downregulated in UUO and TGFβ-stimulated models, and Tan I reversed this downregulation. Inhibition of PGC1α in TGFβ-stimulated cells counteracted the antifibrotic and gluconeogenesis-promoting effects of Tan I. CONCLUSIONS Tanshinone I ameliorated renal fibrosis by enhancing gluconeogenesis through upregulation of PGC1α.
Collapse
MESH Headings
- Disease Models, Animal
- Animals
- Mice
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/pathology
- Fibrosis/drug therapy
- Fibrosis/pathology
- Salvia miltiorrhiza/chemistry
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Antifibrotic Agents/pharmacology
- Antifibrotic Agents/therapeutic use
- Gluconeogenesis/drug effects
- Up-Regulation/drug effects
- Kidney Tubules, Proximal/cytology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Cell Line
- Primary Cell Culture
- Glucose/analysis
- Glucose/metabolism
- Lactic Acid/analysis
- Lactic Acid/metabolism
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/analysis
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors
- Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
- Transforming Growth Factor beta/metabolism
- Mice, Inbred C57BL
- Humans
- Male
Collapse
Affiliation(s)
- Yanfang Bai
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Hui Wen
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Junyan Lin
- The Seventh People’s Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinying Liu
- Department of Rheumatology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Hua Yu
- Shanghai Zhabei District Central Hospital, Shanghai, China
| | - Ming Wu
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Ling Wang
- Department of Nephrology, Shanghai Tenth People’s Hospital of Tongji University, Shanghai, China
| | - Dongping Chen
- Department of Nephrology, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- TCM Institute of Kidney Disease of Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases, Ministry of Education, Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| |
Collapse
|
13
|
Kiyuna LA, Krishnamurthy KA, Homan EB, Langelaar-Makkinje M, Gerding A, Bos T, Oosterhuis D, Overduin RJ, Schreuder AB, de Meijer VE, Olinga P, Derks TGJ, van Eunen K, Bakker BM, Oosterveer MH. Precision-cut liver slices as an ex vivo model to assess impaired hepatic glucose production. Commun Biol 2024; 7:1479. [PMID: 39521914 PMCID: PMC11550398 DOI: 10.1038/s42003-024-07070-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Fasting hypoglycemia is a severe and incompletely understood symptom of various inborn errors of metabolism (IEM). Precision-cut liver slices (PCLS) represent a promising model for studying glucose production ex vivo. This study quantified the net glucose production of human and murine PCLS in the presence of different gluconeogenic precursors. Dihydroxyacetone-supplemented slices from the fed mice yielded the highest rate, further stimulated by forskolin and dibutyryl-cAMP. Moreover, using 13C isotope tracing, we assessed the contribution of glycogenolysis and gluconeogenesis to net glucose production over time. Pharmacological inhibition of the glucose 6-phosphate transporter SLC37A4 markedly reduced net glucose production and increased lactate secretion and glycogen storage, while glucose production was completely abolished in PCLS from glycogen storage disease type Ia and Ib patients. In conclusion, this study identifies PCLS as an effective ex vivo model to study hepatic glucose production and opens opportunities for its future application in IEM research and beyond.
Collapse
Affiliation(s)
- Ligia Akemi Kiyuna
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Esther B Homan
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Miriam Langelaar-Makkinje
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albert Gerding
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Trijnie Bos
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Ruben J Overduin
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Andrea B Schreuder
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Vincent E de Meijer
- Section of HPB Surgery and Liver Transplantation, Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, The Netherlands
| | - Terry G J Derks
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Metabolic Diseases, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Karen van Eunen
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Barbara M Bakker
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Maaike H Oosterveer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
14
|
Hosseini SM, Tingzhu Y, Zaohong R, Ullah F, Liang A, Hua G, Yang L. Regulatory impacts of PPARGC1A gene expression on milk production and cellular metabolism in buffalo mammary epithelial cells. Anim Biotechnol 2024; 35:2344210. [PMID: 38785376 DOI: 10.1080/10495398.2024.2344210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
The PPARGC1A gene plays a fundamental role in regulating cellular energy metabolism, including adaptive thermogenesis, mitochondrial biogenesis, adipogenesis, gluconeogenesis, and glucose/fatty acid metabolism. In a previous study, our group investigated seven SNPs in Mediterranean buffalo associated with milk production traits, and the current study builds on this research by exploring the regulatory influences of the PPARGC1A gene in buffalo mammary epithelial cells (BuMECs). Our findings revealed that knockdown of PPARGC1A gene expression significantly affected the growth of BuMECs, including proliferation, cell cycle, and apoptosis. Additionally, we observed downregulated triglyceride secretion after PPARGC1A knockdown. Furthermore, the critical genes related to milk production, including the STATS, BAD, P53, SREBF1, and XDH genes were upregulated after RNAi, while the FABP3 gene, was downregulated. Moreover, Silencing the PPARGC1A gene led to a significant downregulation of β-casein synthesis in BuMECs. Our study provides evidence of the importance of the PPARGC1A gene in regulating cell growth, lipid, and protein metabolism in the buffalo mammary gland. In light of our previous research, the current study underscores the potential of this gene for improving milk production efficiency and overall dairy productivity in buffalo populations.
Collapse
Affiliation(s)
- Seyed Mahdi Hosseini
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ye Tingzhu
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Ran Zaohong
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Farman Ullah
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Aixin Liang
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Guohua Hua
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Liguo Yang
- National Center for International Research on Animal Genetics, Breeding and Reproduction (NCIRAGBR), College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
15
|
Sauer B, Kueckelhaus J, Lorenz NI, Bozkurt S, Schulte D, Weinem J, Benzarti M, Meiser J, Urban H, Villa G, Harter PN, Münch C, Rieger J, Steinbach JP, Heiland DH, Ronellenfitsch MW. An AMP-activated protein kinase-PGC-1α axis mediates metabolic plasticity in glioblastoma. Clin Transl Med 2024; 14:e70030. [PMID: 39552019 PMCID: PMC11570551 DOI: 10.1002/ctm2.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 09/02/2024] [Accepted: 09/10/2024] [Indexed: 11/19/2024] Open
Abstract
Glioblastoma, the most frequent primary malignant brain tumour in adults, is characterised by profound yet dynamic hypoxia and nutrient depletion. To sustain survival and proliferation, tumour cells are compelled to acquire metabolic plasticity with the induction of adaptive metabolic programs. Here, we interrogated the pathways necessary to enable processing of nutrients other than glucose. We employed genetic approaches (stable/inducible overexpression, CRISPR/Cas9 knockout), pharmacological interventions with a novel inhibitor of AMP-activated protein kinase (AMPK) in glioblastoma cell culture systems and a proteomic approach to investigate mechanisms of metabolic plasticity. Moreover, a spatially resolved multiomic analysis was employed to correlate the gene expression pattern of PGC-1α with the local metabolic and genetic architecture in human glioblastoma tissue sections. A switch from glucose to alternative nutrients triggered an activation of AMPK, which in turn activated PGC-1α-dependent adaptive programs promoting mitochondrial metabolism. This sensor-effector mechanism was essential for metabolic plasticity with both functional AMPK and PGC-1α necessary for survival and growth of cells under nonglucose nutrient sources. In human glioblastoma tissue specimens, PGC-1α-expression correlated with nonhypoxic tumour niches defining a specific metabolic compartment. Our findings reveal a cell-intrinsic nutrient sensing and switching mechanism. The exposure to alternative fuels triggers a starvation signal that subsequently is passed on via AMPK and PGC-1α to induce adaptive programs necessary for broader spectrum nutrient metabolism. The integration of spatially resolved transcriptomic data confirms the relevance of PGC-1α especially in nonhypoxic tumour regions. Thus, the AMPK-PGC-1α axis is a candidate for therapeutic inhibition in glioblastoma. KEY POINTS/HIGHLIGHTS: AMPK activation induces PGC-1α expression in glioblastoma during nutrient scarcity. PGC-1α enables metabolic plasticity by facilitating metabolism of alternative nutrients in glioblastoma. PGC-1α expression is inversely correlated with hypoxic tumour regions in human glioblastomas.
Collapse
Affiliation(s)
- Benedikt Sauer
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Jan Kueckelhaus
- Microenvironment and Immunology Research LaboratoryMedical CenterUniversity of FreiburgFreiburgGermany
- Department of NeurosurgeryMedical CenterUniversity of FreiburgFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of Medicine and Medical CenterComprehensive Cancer Center Freiburg (CCCF)University of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center FreiburgFreiburgGermany
| | - Nadja I. Lorenz
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Süleyman Bozkurt
- Institute of Molecular Systems Medicine, Faculty of MedicineGoethe University FrankfurtFrankfurt am MainGermany
| | - Dorothea Schulte
- Institute of Neurology (Edinger Institute)University Hospital, Goethe University FrankfurtFrankfurtGermany
| | - Jan‐Béla Weinem
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Mohaned Benzarti
- Cancer Metabolism GroupDepartment of Cancer ResearchLuxembourg Institute of HealthLuxembourgLuxembourg
- Faculty of ScienceTechnology and MedicineUniversity of Luxembourg, 2 avenue de UniversitéEsch‐sur‐AlzetteLuxembourg
| | - Johannes Meiser
- Cancer Metabolism GroupDepartment of Cancer ResearchLuxembourg Institute of HealthLuxembourgLuxembourg
- Faculty of ScienceTechnology and MedicineUniversity of Luxembourg, 2 avenue de UniversitéEsch‐sur‐AlzetteLuxembourg
| | - Hans Urban
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Giulia Villa
- Microenvironment and Immunology Research LaboratoryMedical CenterUniversity of FreiburgFreiburgGermany
- Department of NeurosurgeryMedical CenterUniversity of FreiburgFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of Medicine and Medical CenterComprehensive Cancer Center Freiburg (CCCF)University of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center FreiburgFreiburgGermany
| | - Patrick N. Harter
- Institute of Neurology (Edinger Institute)University Hospital, Goethe University FrankfurtFrankfurtGermany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and Ludwig‐Maximilians‐Universität (LMU)MunichGermany
- Center for Neuropathology and Prion Research, Faculty of MedicineLudwig‐Maximilians‐Universität (LMU)MunichGermany
| | - Christian Münch
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- Institute of Molecular Systems Medicine, Faculty of MedicineGoethe University FrankfurtFrankfurt am MainGermany
- Cardio‐Pulmonary InstituteGoethe University FrankfurtFrankfurt am MainGermany
| | - Johannes Rieger
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- Division of Neuro‐OncologyHertie Institute of Clinical Brain ResearchUniversity Hospital TübingenTübingenGermany
| | - Joachim P. Steinbach
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| | - Dieter Henrik Heiland
- Microenvironment and Immunology Research LaboratoryMedical CenterUniversity of FreiburgFreiburgGermany
- Department of NeurosurgeryMedical CenterUniversity of FreiburgFreiburgGermany
- Faculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of Medicine and Medical CenterComprehensive Cancer Center Freiburg (CCCF)University of FreiburgFreiburgGermany
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and University Medical Center FreiburgFreiburgGermany
| | - Michael W. Ronellenfitsch
- Dr. Senckenberg Institute of NeurooncologyUniversity HospitalGoethe University FrankfurtFrankfurt am MainGermany
- University Cancer Center Frankfurt (UCT), University HospitalGoethe University FrankfurtFrankfurt am MainGermany
- German Cancer Consortium (DKTK), partner site Frankfurt, a partnership between DKFZ and University Hospital FrankfurtGoethe University FrankfurtFrankfurt am MainGermany
- Frankfurt Cancer Institute (FCI)University HospitalGoethe University FrankfurtFrankfurt am MainGermany
| |
Collapse
|
16
|
Caldovic L, Ahn JJ, Andricovic J, Balick VM, Brayer M, Chansky PA, Dawson T, Edwards AC, Felsen SE, Ismat K, Jagannathan SV, Mann BT, Medina JA, Morizono T, Morizono M, Salameh S, Vashist N, Williams EC, Zhou Z, Morizono H. Datamining approaches for examining the low prevalence of N-acetylglutamate synthase deficiency and understanding transcriptional regulation of urea cycle genes. J Inherit Metab Dis 2024; 47:1175-1193. [PMID: 37847851 PMCID: PMC11586597 DOI: 10.1002/jimd.12687] [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: 05/30/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
Ammonia, which is toxic to the brain, is converted into non-toxic urea, through a pathway of six enzymatically catalyzed steps known as the urea cycle. In this pathway, N-acetylglutamate synthase (NAGS, EC 2.3.1.1) catalyzes the formation of N-acetylglutamate (NAG) from glutamate and acetyl coenzyme A. NAGS deficiency (NAGSD) is the rarest of the urea cycle disorders, yet is unique in that ureagenesis can be restored with the drug N-carbamylglutamate (NCG). We investigated whether the rarity of NAGSD could be due to low sequence variation in the NAGS genomic region, high NAGS tolerance for amino acid replacements, and alternative sources of NAG and NCG in the body. We also evaluated whether the small genomic footprint of the NAGS catalytic domain might play a role. The small number of patients diagnosed with NAGSD could result from the absence of specific disease biomarkers and/or short NAGS catalytic domain. We screened for sequence variants in NAGS regulatory regions in patients suspected of having NAGSD and found a novel NAGS regulatory element in the first intron of the NAGS gene. We applied the same datamining approach to identify regulatory elements in the remaining urea cycle genes. In addition to the known promoters and enhancers of each gene, we identified several novel regulatory elements in their upstream regions and first introns. The identification of cis-regulatory elements of urea cycle genes and their associated transcription factors holds promise for uncovering shared mechanisms governing urea cycle gene expression and potentially leading to new treatments for urea cycle disorders.
Collapse
Affiliation(s)
- Ljubica Caldovic
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Julie J. Ahn
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Jacklyn Andricovic
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Veronica M. Balick
- Department of Biochemistry and Molecular MedicineThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Mallory Brayer
- Department of Biological SciencesThe George Washington UniversityWashingtonDCUSA
| | - Pamela A. Chansky
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Tyson Dawson
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- AMPEL BioSolutions LLCCharlottesvilleVirginiaUSA
| | - Alex C. Edwards
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Center for Neuroscience ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
| | - Sara E. Felsen
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Center for Neuroscience ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
| | - Karim Ismat
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Sveta V. Jagannathan
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Brendan T. Mann
- Department of Microbiology, Immunology, and Tropical MedicineSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Jacob A. Medina
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Toshio Morizono
- College of Science and EngineeringUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Michio Morizono
- College of Science and EngineeringUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Shatha Salameh
- Department of Pharmacology & PhysiologySchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Sheikh Zayed Institute for Pediatric Surgical InnovationChildren's National HospitalWashingtonDCUSA
| | - Neerja Vashist
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Emily C. Williams
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
- The George Washington University Cancer Center, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDCUSA
| | - Zhe Zhou
- Department of Civil and Environmental EngineeringThe George Washington UniversityWashingtonDCUSA
| | - Hiroki Morizono
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| |
Collapse
|
17
|
Mutlu B, Sharabi K, Sohn JH, Yuan B, Latorre-Muro P, Qin X, Yook JS, Lin H, Yu D, Camporez JPG, Kajimura S, Shulman GI, Hui S, Kamenecka TM, Griffin PR, Puigserver P. Small molecules targeting selective PCK1 and PGC-1α lysine acetylation cause anti-diabetic action through increased lactate oxidation. Cell Chem Biol 2024; 31:1772-1786.e5. [PMID: 39341205 PMCID: PMC11500315 DOI: 10.1016/j.chembiol.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/27/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024]
Abstract
Small molecules selectively inducing peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α acetylation and inhibiting glucagon-dependent gluconeogenesis causing anti-diabetic effects have been identified. However, how these small molecules selectively suppress the conversion of gluconeogenic metabolites into glucose without interfering with lipogenesis is unknown. Here, we show that a small molecule SR18292 inhibits hepatic glucose production by increasing lactate and glucose oxidation. SR18292 increases phosphoenolpyruvate carboxykinase 1 (PCK1) acetylation, which reverses its gluconeogenic reaction and favors oxaloacetate (OAA) synthesis from phosphoenolpyruvate. PCK1 reverse catalytic reaction induced by SR18292 supplies OAA to tricarboxylic acid (TCA) cycle and is required for increasing glucose and lactate oxidation and suppressing gluconeogenesis. Acetylation mimetic mutant PCK1 K91Q favors anaplerotic reaction and mimics the metabolic effects of SR18292 in hepatocytes. Liver-specific expression of PCK1 K91Q mutant ameliorates hyperglycemia in obese mice. Thus, SR18292 blocks gluconeogenesis by enhancing gluconeogenic substrate oxidation through PCK1 lysine acetylation, supporting the anti-diabetic effects of these small molecules.
Collapse
Affiliation(s)
- Beste Mutlu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA; Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jee Hyung Sohn
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Bo Yuan
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Xin Qin
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jin-Seon Yook
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Hua Lin
- Department of Molecular Medicine, The Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, Jupiter, FL 33458, USA
| | - Deyang Yu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - João Paulo G Camporez
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520-8020, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520-8020, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Chevy Chase, MD 020815, USA
| | - Gerald I Shulman
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06520-8020, USA; Department of Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520-8020, USA; Howard Hughes Medical Institute, Chevy Chase, MD 020815, USA
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, 655 Huntington Avenue, Boston, MA 02115, USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, Jupiter, FL 33458, USA
| | - Patrick R Griffin
- Department of Molecular Medicine, The Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, Jupiter, FL 33458, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
18
|
Kositanurit W, Siritaweechai N, Varachotisate P, Burana C, Sukswai N, Surintrspanont J, Siriviriyakul P, Kaikaew K, Werawatganon D. Genistein mitigates diet-induced obesity and metabolic dysfunctions in gonadectomized mice with some sex-differential effects. Front Endocrinol (Lausanne) 2024; 15:1392866. [PMID: 39351533 PMCID: PMC11439649 DOI: 10.3389/fendo.2024.1392866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 08/26/2024] [Indexed: 10/04/2024] Open
Abstract
Background Obesity is associated with insulin resistance (IR) and metabolic dysfunction-associated steatotic liver disease (MASLD). Genistein, an isoflavone, is a promising natural compound for preventing and treating obesity and metabolic dysfunctions. We aimed to investigate the sex-specific protective effects of genistein on obesity, IR, and MASLD in a murine model of sex hormone deprivation with diet-induced obesity (DIO), mimicking postmenopausal women or aging men with metabolic syndrome. Methods Gonadectomized and sham-operated C57BL/6NJcl mice were fed a high-fat high-sucrose diet for 4 weeks to induce obesity (7 mice per group). In gonadectomized mice, genistein (16 mg/kg/day) or vehicle (7.5% dimethyl sulfoxide) was orally administered for 45 days. We assessed glucose homeostasis parameters, hepatic histopathology, and hepatic gene expression to investigate the effects of gonadectomy and genistein treatment. Results Gonadectomy exacerbated adiposity in both sexes. Ovariectomy diminished the protective effects of female gonadal hormones on the homeostatic model assessment for insulin resistance (HOMA-IR), serum alanine transaminase levels, hepatic steatosis score, and the expression of hepatic genes associated with MASLD progression and IR, such as Fasn, Srebf1, Saa1, Cd36, Col1a1, Pck1, and Ppargc1a. Genistein treatment in gonadectomized mice significantly reduced body weight gain and the hepatic steatosis score in both sexes. However, genistein treatment significantly attenuated HOMA-IR and the expression of the hepatic genes only in female mice. Conclusion Genistein treatment mitigates DIO-related MASLD in both male and female gonadectomized mice. Regarding hepatic gene expression associated with MASLD and IR, the beneficial effect of genistein was significantly evident only in female mice. This study suggests a potential alternative application of genistein in individuals with obesity and sex hormone deprivation, yet pending clinical trials.
Collapse
Affiliation(s)
- Weerapat Kositanurit
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Natakorn Siritaweechai
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pachara Varachotisate
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Chuti Burana
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Narittee Sukswai
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Jerasit Surintrspanont
- Precision Pathology of Neoplasia Research Group, Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Pathology, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok, Thailand
| | - Prasong Siriviriyakul
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Kasiphak Kaikaew
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| | - Duangporn Werawatganon
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Alternative and Complementary Medicine for Gastrointestinal and Liver Diseases, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
19
|
da Costa CS, de Oliveira TF, Dos Santos FCF, Padilha AS, Krause M, Carneiro MTWD, Miranda-Alves L, Graceli JB. Subacute cadmium exposure changes different metabolic functions, leading to type 1 and 2 diabetes mellitus features in female rats. ENVIRONMENTAL TOXICOLOGY 2024; 39:4278-4297. [PMID: 38712533 DOI: 10.1002/tox.24306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/15/2024] [Accepted: 04/23/2024] [Indexed: 05/08/2024]
Abstract
Cadmium (Cd) is a heavy metal that acts as endocrine disrupting chemical (EDC). Few studies have investigated the effects of Cd exposure on metabolic dysfunctions, such as type 1 and 2 diabetes mellitus (T1DM and T2DM). Thus, we assessed whether subacute Cd exposure at occupational levels causes abnormalities in white adipose tissue (WAT), liver, pancreas, and skeletal muscle. We administered cadmium chloride (CdCl2) (100 ppm in drinking water for 30 days) to female rats and evaluated Cd levels in serum and metabolic organs, morphophysiology, inflammation, oxidative stress, fibrosis, and gene expression. High Cd levels were found in serum, WAT, liver, pancreas, and skeletal muscle. Cd-exposed rats showed low adiposity, dyslipidemia, insulin resistance, systemic inflammation, and oxidative stress compared to controls. Cd exposure reduced adipocyte size, hyperleptinemia, increased cholesterol levels, inflammation, apoptosis and fibrosis in WAT. Cd-exposed rats had increased liver cholesterol levels, insulin receptor beta (IRβ) and peroxisome proliferator-activated receptor-gamma coactivator-1alpha (PGC1α) expression, karyomegaly, inflammation, and fibrosis. Cd exposure reduced insulin levels and pancreatic islet size and increased inflammation and fibrosis. Cd exposure reduced skeletal muscle fiber diameter and increased IR expression and inflammation. Finally, strong positive correlations were observed between serum, tissue Cd levels, abnormal morphology, tissue inflammation and fibrosis. Thus, these data suggest that subacute Cd exposure impairs WAT, liver, pancreas and skeletal muscle function, leading to T1DM and T2DM features and other complications in female rats.
Collapse
Affiliation(s)
- Charles S da Costa
- Department of Morphology, Federal University of Espirito Santo, Vitória, Brazil
| | | | | | | | - Maiara Krause
- Department of Chemistry, Federal University of Espirito Santo, Vitória, Brazil
| | | | - Leandro Miranda-Alves
- Experimental Endocrinology Research, Development and Innovation Group, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jones B Graceli
- Department of Morphology, Federal University of Espirito Santo, Vitória, Brazil
| |
Collapse
|
20
|
Louvrou V, Solianik R, Brazaitis M, Erhardt S. Exploring the effect of prolonged fasting on kynurenine pathway metabolites and stress markers in healthy male individuals. Eur J Clin Nutr 2024; 78:677-683. [PMID: 38789718 PMCID: PMC11300305 DOI: 10.1038/s41430-024-01451-7] [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: 12/07/2023] [Accepted: 05/08/2024] [Indexed: 05/26/2024]
Abstract
BACKGROUND/OBJECTIVES Prolonged fasting triggers a stress response within the human body. Our objective was to investigate the impact of prolonged fasting, in conjunction with stress, on kynurenine pathway metabolites. SUBJECTS/METHODS Healthy males were divided into fasting group (zero-calorie-restriction) for 6 days (FAST, n = 14), and control group (CON, n = 10). Blood and saliva samples were collected at baseline, Day 2, Day 4, Day 6 during fasting period, and 1 week after resuming regular diet. Plasma levels of kynurenine pathway metabolites were measured using ultra-performance liquid chromatography-mass spectrometry (UPLC-MS/MS). Plasma and salivary samples were analyzed for stress markers. RESULTS A pronounced activation of the kynurenine pathway in individuals on FAST trial was revealed. Concentrations of picolinic acid (PIC), kynurenic acid (KYNA) and 3-hydroxykynurenine (3-HK) were significantly increased, with peak levels observed on Day 6 (P < 0.0001). Conversely, concentrations of tryptophan (TRP) and quinolinic acid (QUIN) decreased (P < 0.0001), while kynurenine (KYN) and nicotinamide (NAM) levels remained stable. Cortisol and noradrenaline concentrations remained unchanged. However, adrenaline levels significantly increased on Day 4 within FAST compared to CON (P = 0.005). Notably, all deviations in kynurenine pathway metabolite levels returned to baseline values upon resuming regular diet following the 6-day fasting regimen, even when weight and BMI parameters were not restored. CONCLUSIONS Extended fasting over 6 days induces the kynurenine pathway and has minimal effects on stress markers. Restoration of metabolite concentrations upon regular feeding implies rapid adaptation of the kynurenine pathway synthetic enzymes to maintain homeostasis when faced with perturbations.
Collapse
Affiliation(s)
- Varvara Louvrou
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Rima Solianik
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Marius Brazaitis
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Sophie Erhardt
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania.
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
21
|
Locke B, Campbell E, Lu R. CREB3 mediates the transcriptional regulation of PGC-1α, a master regulator of energy homeostasis and mitochondrial biogenesis. FEBS Lett 2024; 598:1730-1739. [PMID: 38697949 DOI: 10.1002/1873-3468.14897] [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: 12/13/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/05/2024]
Abstract
Lipid metabolism hinges on a balance between lipogenesis and fatty acid oxidation (FAO). Disruptions in this balance can induce endoplasmic reticulum (ER) stress triggering the unfolded protein response (UPR) and contribute to metabolic diseases. The UPR protein, Luman or CREB3, has recently been implicated in metabolic regulation-CREB3 knockout mice exhibit resistance to diet-induced obesity and altered insulin sensitivity. Here, we show that CREB3 activated PPARGC1A transcription from a 1 kb promoter region. An increase in CREB3 expression correlated inversely with endogenous PPARGC1A mRNA levels and genes involved in FAO. As PGC-1α encoded by PPARGC1A is a master regulator of mitochondrial biogenesis and energy homeostasis, these findings demonstrate that CREB3 is a transcriptional regulator of PGC-1α, underlining the potential role of CREB3 in energy metabolism.
Collapse
Affiliation(s)
- Briana Locke
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Elena Campbell
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| | - Ray Lu
- Department of Molecular and Cellular Biology, University of Guelph, Canada
| |
Collapse
|
22
|
Desmet SJ, Thommis J, Vanderhaeghen T, Vandenboorn EMF, Clarisse D, Li Y, Timmermans S, Fijalkowska D, Ratman D, Van Hamme E, De Cauwer L, Staels B, Brunsveld L, Peelman F, Libert C, Tavernier J, De Bosscher K. Crosstalk interactions between transcription factors ERRα and PPARα assist PPARα-mediated gene expression. Mol Metab 2024; 84:101938. [PMID: 38631478 PMCID: PMC11059514 DOI: 10.1016/j.molmet.2024.101938] [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] [Received: 01/15/2024] [Revised: 03/10/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024] Open
Abstract
OBJECTIVE The peroxisome proliferator-activated receptor α (PPARα) is a transcription factor driving target genes involved in fatty acid β-oxidation. To what extent various PPARα interacting proteins may assist its function as a transcription factor is incompletely understood. An ORFeome-wide unbiased mammalian protein-protein interaction trap (MAPPIT) using PPARα as bait revealed a PPARα-ligand-dependent interaction with the orphan nuclear receptor estrogen-related receptor α (ERRα). The goal of this study was to characterize the nature of the interaction in depth and to explore whether it was of physiological relevance. METHODS We used orthogonal protein-protein interaction assays and pharmacological inhibitors of ERRα in various systems to confirm a functional interaction and study the impact of crosstalk mechanisms. To characterize the interaction surfaces and contact points we applied a random mutagenesis screen and structural overlays. We pinpointed the extent of reciprocal ligand effects of both nuclear receptors via coregulator peptide recruitment assays. On PPARα targets revealed from a genome-wide transcriptome analysis, we performed an ERRα chromatin immunoprecipitation analysis on both fast and fed mouse livers. RESULTS Random mutagenesis scanning of PPARα's ligand-binding domain and coregulator profiling experiments supported the involvement of (a) bridging coregulator(s), while recapitulation of the interaction in vitro indicated the possibility of a trimeric interaction with RXRα. The PPARα·ERRα interaction depends on 3 C-terminal residues within helix 12 of ERRα and is strengthened by both PGC1α and serum deprivation. Pharmacological inhibition of ERRα decreased the interaction of ERRα to ligand-activated PPARα and revealed a transcriptome in line with enhanced mRNA expression of prototypical PPARα target genes, suggesting a role for ERRα as a transcriptional repressor. Strikingly, on other PPARα targets, including the isolated PDK4 enhancer, ERRα behaved oppositely. Chromatin immunoprecipitation analyses demonstrate a PPARα ligand-dependent ERRα recruitment onto chromatin at PPARα-binding regions, which is lost following ERRα inhibition in fed mouse livers. CONCLUSIONS Our data support the coexistence of multiple layers of transcriptional crosstalk mechanisms between PPARα and ERRα, which may serve to finetune the activity of PPARα as a nutrient-sensing transcription factor.
Collapse
Affiliation(s)
- Sofie J Desmet
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Jonathan Thommis
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Tineke Vanderhaeghen
- VIB Center for Inflammation Research, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Edmee M F Vandenboorn
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, the Netherlands
| | - Dorien Clarisse
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Yunkun Li
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Steven Timmermans
- VIB Center for Inflammation Research, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Daria Fijalkowska
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Dariusz Ratman
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | | | - Lode De Cauwer
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Bart Staels
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Luc Brunsveld
- Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, 5612AZ Eindhoven, the Netherlands
| | - Frank Peelman
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Claude Libert
- VIB Center for Inflammation Research, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Jan Tavernier
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium
| | - Karolien De Bosscher
- VIB Center for Medical Biotechnology, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
23
|
Samson N, Bosoi CR, Roy C, Turcotte L, Tribouillard L, Mouchiroud M, Berthiaume L, Trottier J, Silva HCG, Guerbette T, Plata-Gómez AB, Besse-Patin A, Montoni A, Ilacqua N, Lamothe J, Citron YR, Gélinas Y, Gobeil S, Zoncu R, Caron A, Morissette M, Pellegrini L, Rochette PJ, Estall JL, Efeyan A, Shum M, Audet-Walsh É, Barbier O, Marette A, Laplante M. HSDL2 links nutritional cues to bile acid and cholesterol homeostasis. SCIENCE ADVANCES 2024; 10:eadk9681. [PMID: 38820148 PMCID: PMC11141617 DOI: 10.1126/sciadv.adk9681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 04/29/2024] [Indexed: 06/02/2024]
Abstract
In response to energy and nutrient shortage, the liver triggers several catabolic processes to promote survival. Despite recent progress, the precise molecular mechanisms regulating the hepatic adaptation to fasting remain incompletely characterized. Here, we report the identification of hydroxysteroid dehydrogenase-like 2 (HSDL2) as a mitochondrial protein highly induced by fasting. We show that the activation of PGC1α-PPARα and the inhibition of the PI3K-mTORC1 axis stimulate HSDL2 expression in hepatocytes. We found that HSDL2 depletion decreases cholesterol conversion to bile acids (BAs) and impairs FXR activity. HSDL2 knockdown also reduces mitochondrial respiration, fatty acid oxidation, and TCA cycle activity. Bioinformatics analyses revealed that hepatic Hsdl2 expression positively associates with the postprandial excursion of various BA species in mice. We show that liver-specific HSDL2 depletion affects BA metabolism and decreases circulating cholesterol levels upon refeeding. Overall, our report identifies HSDL2 as a fasting-induced mitochondrial protein that links nutritional signals to BAs and cholesterol homeostasis.
Collapse
Affiliation(s)
- Nolwenn Samson
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Cristina R. Bosoi
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Christian Roy
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Laurie Turcotte
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Laura Tribouillard
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Mathilde Mouchiroud
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
| | - Line Berthiaume
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
| | - Jocelyn Trottier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
| | - Heitor C. G. Silva
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Thomas Guerbette
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Ana Belén Plata-Gómez
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Aurèle Besse-Patin
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
| | - Alicia Montoni
- Axe Médecine régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Hôpital du Saint-Sacrement, Québec, QC, Canada
| | - Nicolò Ilacqua
- Faculté de médecine, Université Laval, Québec, QC, Canada
- Centre de recherche CERVO, Québec, QC, Canada
| | - Jennifer Lamothe
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | - Yemima R. Citron
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA, USA
| | - Yves Gélinas
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
| | | | - Roberto Zoncu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Innovative Genomics Initiative at the University of California, Berkeley, Berkeley, CA, USA
| | - Alexandre Caron
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - Mathieu Morissette
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Luca Pellegrini
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, QC, Canada
| | - Patrick J. Rochette
- Faculté de médecine, Université Laval, Québec, QC, Canada
- Axe Médecine régénératrice, Centre de Recherche du CHU de Québec-Université Laval, Hôpital du Saint-Sacrement, Québec, QC, Canada
- Département d’Ophtalmologie et ORL – chirurgie cervico-faciale, Université Laval, Québec, QC, Canada
| | - Jennifer L. Estall
- Institut de recherches cliniques de Montréal (IRCM), Montréal, QC, Canada
- Faculté de médecine, Université de Montréal, Montréal, QC, Canada
| | - Alejo Efeyan
- Metabolism and Cell Signaling Laboratory, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Michael Shum
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Étienne Audet-Walsh
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Olivier Barbier
- Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec-Université Laval, Axe Endocrinologie et néphrologie, Québec, QC, Canada
- Faculté de Pharmacie, Université Laval, Québec, QC, Canada
| | - André Marette
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| | - Mathieu Laplante
- Centre de Recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, QC, Canada
- Centre de recherche sur le cancer de l’Université Laval, Université Laval, Québec, QC, Canada
- Faculté de médecine, Université Laval, Québec, QC, Canada
| |
Collapse
|
24
|
Yu Q, Zuo X, Bai H, Zhang S, Luan J, Zhao Q, Zhao X, Feng X. Alleviative effects of the parthenolide derivative ACT001 on insulin resistance induced by sodium propionate combined with a high-fat diet and its potential mechanisms. Eur J Pharmacol 2024; 971:176529. [PMID: 38554931 DOI: 10.1016/j.ejphar.2024.176529] [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: 10/29/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/02/2024]
Abstract
The increasing side effects of traditional medications used to treat type II diabetes have made research into the development of safer and more effective natural medications necessary. ACT001, a derivative of parthenolide, has been shown to have good anti-inflammatory and antitumor effects; however, its role in diabetes is unclear. The short-chain fatty acid propionate is a common food preservative that has been found to cause disturbances in glucose metabolism in mice and humans. This study aimed to investigate whether sodium propionate could aggravate insulin resistance in obese mice and cause diabetes and to study the alleviative effects and potential mechanisms of action of ACT001 on insulin resistance in diabetic mice. Type II diabetic mice were adminietered sodium propionate combined with a high-fat diet (HFD + propionate) by gavage daily for four weeks. Biochemical analysis showed that ACT001 significantly affected blood glucose concentration in diabetic mice, mainly by downregulating the expression of phosphoenolpyruvate carboxykinase 2 and glucose-6-phosphatase. Meanwhile, the level of fatty acid-binding protein 4 in the liver was significantly decreased. ACT001 has a protective effect on the liver and adipose tissue of mice. In addition, the results of the running wheel experiment indicated that ACT001 alleviated the circadian rhythm disorder caused by insulin resistance to a certain extent. This study revealed the potential mechanism by which ACT001 alleviates insulin resistance and provides ideas for developing natural antidiabetic drugs.
Collapse
Affiliation(s)
- Qian Yu
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Xiang Zuo
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Huijuan Bai
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Shuhui Zhang
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Jialu Luan
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China
| | - Qili Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Xin Zhao
- Institute of Robotics & Automatic Information System, College of Artificial Intelligence, Nankai University, Tianjin, 300071, China
| | - Xizeng Feng
- College of Life Science, State Key Laboratory of Medicinal Chemical Biology, The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
25
|
Spinelli S, Bruschi M, Passalacqua M, Guida L, Magnone M, Sturla L, Zocchi E. Estrogen-Related Receptor α: A Key Transcription Factor in the Regulation of Energy Metabolism at an Organismic Level and a Target of the ABA/LANCL Hormone Receptor System. Int J Mol Sci 2024; 25:4796. [PMID: 38732013 PMCID: PMC11084903 DOI: 10.3390/ijms25094796] [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: 03/25/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
The orphan nuclear receptor ERRα is the most extensively researched member of the estrogen-related receptor family and holds a pivotal role in various functions associated with energy metabolism, especially in tissues characterized by high energy requirements, such as the heart, skeletal muscle, adipose tissue, kidney, and brain. Abscisic acid (ABA), traditionally acknowledged as a plant stress hormone, is detected and actively functions in organisms beyond the land plant kingdom, encompassing cyanobacteria, fungi, algae, protozoan parasites, lower Metazoa, and mammals. Its ancient, cross-kingdom role enables ABA and its signaling pathway to regulate cell responses to environmental stimuli in various organisms, such as marine sponges, higher plants, and humans. Recent advancements in understanding the physiological function of ABA and its mammalian receptors in governing energy metabolism and mitochondrial function in myocytes, adipocytes, and neuronal cells suggest potential therapeutic applications for ABA in pre-diabetes, diabetes, and cardio-/neuroprotection. The ABA/LANCL1-2 hormone/receptor system emerges as a novel regulator of ERRα expression levels and transcriptional activity, mediated through the AMPK/SIRT1/PGC-1α axis. There exists a reciprocal feed-forward transcriptional relationship between the LANCL proteins and transcriptional coactivators ERRα/PGC-1α, which may be leveraged using natural or synthetic LANCL agonists to enhance mitochondrial function across various clinical contexts.
Collapse
Affiliation(s)
- Sonia Spinelli
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, Via Gerolamo Gaslini 5, 16147 Genova, Italy
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (M.P.); (L.G.); (M.M.); (L.S.)
| | - Mario Passalacqua
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (M.P.); (L.G.); (M.M.); (L.S.)
| | - Lucrezia Guida
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (M.P.); (L.G.); (M.M.); (L.S.)
| | - Mirko Magnone
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (M.P.); (L.G.); (M.M.); (L.S.)
| | - Laura Sturla
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (M.P.); (L.G.); (M.M.); (L.S.)
| | - Elena Zocchi
- Section Biochemistry, Department of Experimental Medicine (DIMES), University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy; (M.P.); (L.G.); (M.M.); (L.S.)
| |
Collapse
|
26
|
Chen M, Zhu J, Luo H, Mu W, Guo L. The journey towards physiology and pathology: Tracing the path of neuregulin 4. Genes Dis 2024; 11:687-700. [PMID: 37692526 PMCID: PMC10491916 DOI: 10.1016/j.gendis.2023.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 02/11/2023] [Accepted: 03/05/2023] [Indexed: 09/12/2023] Open
Abstract
Neuregulin 4 (Nrg4), an epidermal growth factor (EGF) family member, can bind to and activate the ErbB4 receptor tyrosine kinase. Nrg4 has five different isoforms by alternative splicing and performs a wide variety of functions. Nrg4 is involved in a spectrum of physiological processes including neurobiogenesis, lipid metabolism, glucose metabolism, thermogenesis, and angiogenesis. In pathological processes, Nrg4 inhibits inflammatory factor levels and suppresses apoptosis in inflammatory diseases. In addition, Nrg4 could ameliorate obesity, insulin resistance, and cardiovascular diseases. Furthermore, Nrg4 improves non-alcoholic fatty liver disease (NAFLD) by promoting autophagy, improving lipid metabolism, and inhibiting cell death of hepatocytes. Besides, Nrg4 is closely related to the development of cancer, hyperthyroidism, and some other diseases. Therefore, elucidation of the functional role and mechanisms of Nrg4 will provide a clearer view of the therapeutic potential and possible risks of Nrg4.
Collapse
Affiliation(s)
- Min Chen
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Jieying Zhu
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Hongyang Luo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Wangjing Mu
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Liang Guo
- School of Exercise and Health and Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
27
|
Shin MK, Tang WY, Amorim MR, Sham JSK, Polotsky VY. Carotid body denervation improves hyperglycemia in obese mice. J Appl Physiol (1985) 2024; 136:233-243. [PMID: 38126089 PMCID: PMC11219014 DOI: 10.1152/japplphysiol.00215.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/14/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023] Open
Abstract
The carotid bodies (CBs) have been implicated in glucose abnormalities in obesity via elevation of activity of the sympathetic nervous system. Obesity-induced hypertension is mediated by insulin receptor (INSR) signaling and by leptin, which binds to the leptin receptor (LEPRb) in CB and activates transient receptor potential channel subfamily M member 7 (TRPM7). We hypothesize that in mice with diet-induced obesity, hyperglycemia, glucose intolerance, and insulin resistance will be attenuated by the CB denervation (carotid sinus nerve dissection, CSND) and by knockdown of Leprb, Trpm7, and Insr gene expression in CB. In series of experiments in 75 male diet-induced obese (DIO) mice, we performed either CSND (vs. sham) surgeries or shRNA-induced suppression of Leprb, Trpm7, or Insr gene expression in CB, followed by blood pressure telemetry, intraperitoneal glucose tolerance and insulin tolerance tests, and measurements of fasting plasma insulin, leptin, corticosterone, glucagon and free fatty acids (FFAs) levels, hepatic expression of gluconeogenesis enzymes phosphoenolpyruvate carboxykinase (PEPCK) and glucose 6-phosphatase (G-6-Pase) mRNA and liver glycogen levels. CSND decreased blood pressure, fasting blood glucose levels and improved glucose tolerance without any effect on insulin resistance. CSND did not affect any hormone levels and gluconeogenesis enzymes, but increased liver glycogen level. Genetic knockdown of CB Leprb, Trpm7, and Insr had no effect on glucose metabolism. We conclude that CB contributes to hyperglycemia of obesity, probably by modulation of the glycogen-glucose equilibrium. Diabetogenic effects of obesity on CB in mice do not occur via activation of CB Leprb, Trpm7, and Insr.NEW & NOTEWORTHY This paper provides first evidence that carotid body denervation abolishes hypertension and improves fasting blood glucose levels and glucose tolerance in mice with diet-induced obesity. Furthermore, we have shown that this phenomenon is associated with increased liver glycogen content, whereas insulin sensitivity and enzymes of gluconeogenesis were not affected.
Collapse
Affiliation(s)
- Mi-Kyung Shin
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - Wan-Yee Tang
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States
| | - Mateus R Amorim
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| | - James S-K Sham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Vsevolod Y Polotsky
- Department of Anesthesiology and Critical Care Medicine, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
- Department of Pharmacology and Physiology, George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, United States
| |
Collapse
|
28
|
Yasuda M. [Acute hepatic porphyrias: pathophysiology and pathogenesis of acute attacks]. Rinsho Shinkeigaku 2024; 64:8-16. [PMID: 38092415 DOI: 10.5692/clinicalneurol.cn-001856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Heme is an iron-containing molecule essential for virtually all living organisms. However, excessive heme is cytotoxic, necessitating tight regulation of intracellular heme concentration. The acute hepatic porphyrias (AHPs) are a group of rare inborn errors of heme biosynthesis that are characterized by episodic acute neurovisceral attacks that are precipitated by various factors. The AHPs are often misdiagnosed, as the acute attack symptom are non-specific and can be attributed to other more common causes. Understanding how heme biosynthesis is dysregulated in AHP patients and the mechanism by which acute attacks are precipitated will aid in accurate and rapid diagnoses, and subsequently, appropriate treatment of these disorders. Therefore, this review article will focus on the biochemical and molecular changes that occur during an acute attack and present what is currently known regarding the underlying pathogenesis of acute attacks.
Collapse
Affiliation(s)
- Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai
| |
Collapse
|
29
|
Wang GY, Qin SL, Zheng YN, Geng HJ, Chen L, Yao JH, Deng L. Propionate promotes gluconeogenesis by regulating mechanistic target of rapamycin (mTOR) pathway in calf hepatocytes. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2023; 15:88-98. [PMID: 37841648 PMCID: PMC10568569 DOI: 10.1016/j.aninu.2023.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 06/13/2023] [Accepted: 07/18/2023] [Indexed: 10/17/2023]
Abstract
Enhancing hepatic gluconeogenesis is one of the main modes of meeting the glucose requirement of dairy cows. This study attempted to determine whether the gluconeogenesis precursor propionate had an effect on the expression of the main genes involved in gluconeogenesis in calf hepatocytes and elucidate the associated mechanisms. Calf hepatocytes were obtained from 5 healthy calves (1 d old; 30 to 40 kg) and exposed to 0-, 1-, 2.5-, or 5-mM sodium propionate (NaP), which is known to promote the expression of genes involved in the gluconeogenesis pathway, including fructose 1,6-bisphosphatase, phosphoenolpyruvate carboxykinase, and glucose-6-phosphatase. With regard to the underlying mechanism, propionate promoted the expression of peroxisome proliferator-activated receptor gamma coactivator 1-alpha, hepatocyte nuclear factor 4, and forkhead box O1 (transcription factors that regulate the expression of hepatic gluconeogenic genes) by promoting mammalian target of rapamycin complex 1 (mTORC1), but inhibiting mTORC2 activity (P < 0.01). We also established a model of palmitic acid (PA)-induced hepatic injury in calf hepatocytes and found that PA could inhibit the gluconeogenic capacity of calf hepatocytes by suppressing the expression of gluconeogenic genes, inhibiting mTORC1, and promoting the activity of mTORC2 (P < 0.01). In contrast, NaP provided protection to calf hepatocytes by counteracting the inhibitory effect of PA on the gluconeogenic capacity of calf hepatocytes (P < 0.05). Collectively, these findings indicate that NaP enhances the gluconeogenic capacity of calf hepatocytes by regulating the mTOR pathway activity. Thus, in addition to improving the glucose production potential, propionate may have therapeutic potential for the treatment of hepatic injury in dairy cows.
Collapse
Affiliation(s)
| | | | - Yi Ning Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Hui Jun Geng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lei Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Jun Hu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
30
|
Ferreira B, Heredia A, Serpa J. An integrative view on glucagon function and putative role in the progression of pancreatic neuroendocrine tumours (pNETs) and hepatocellular carcinomas (HCC). Mol Cell Endocrinol 2023; 578:112063. [PMID: 37678603 DOI: 10.1016/j.mce.2023.112063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/16/2023] [Accepted: 09/02/2023] [Indexed: 09/09/2023]
Abstract
Cancer metabolism research area evolved greatly, however, is still unknown the impact of systemic metabolism control and diet on cancer. It makes sense that systemic regulators of metabolism can act directly on cancer cells and activate signalling, prompting metabolic remodelling needed to sustain cancer cell survival, tumour growth and disease progression. In the present review, we describe the main glucagon functions in the control of glycaemia and of metabolic pathways overall. Furthermore, an integrative view on how glucagon and related signalling pathways can contribute for pancreatic neuroendocrine tumours (pNETs) and hepatocellular carcinomas (HCC) progression, since pancreas and liver are the major organs exposed to higher levels of glucagon, pancreas as a producer and liver as a scavenger. The main objective is to bring to discussion some glucagon-dependent mechanisms by presenting an integrative view on microenvironmental and systemic aspects in pNETs and HCC biology.
Collapse
Affiliation(s)
- Bárbara Ferreira
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal
| | - Adrián Heredia
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal; Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz MB, 1649-028, Lisboa, Portugal
| | - Jacinta Serpa
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo Dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal; Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal.
| |
Collapse
|
31
|
Li D, Ye C, Liu P, Sun T, Qin Y, Wan X. PGC1α deficiency reverses cholestasis-induced liver injury via attenuating hepatic inflammation and promoting bile duct remodeling. Acta Histochem 2023; 125:152097. [PMID: 37813066 DOI: 10.1016/j.acthis.2023.152097] [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: 05/08/2023] [Revised: 09/06/2023] [Accepted: 09/30/2023] [Indexed: 10/11/2023]
Abstract
OBJECTIVES Cholestatic liver diseases are characterized by hepatocellular damage, cholangiocyte proliferation, and progressive fibrosis. Bile duct ligation (BDL) is widely used to resemble liver injuries induced by cholestasis. Peroxisome proliferator-activated receptor-gamma coactivator 1 alpha (PGC1α) was reported to play a critical role in multiple biological responses. Nevertheless, whether PGC1α is involved in bile acid metabolism and biliary disorders remains unclear. This study aimed to investigate the effect of PGC1α on hepatic responses after cholestatic injury. MATERIALS AND METHODS Wild-type mice were subjected to BDL or sham surgery for 14 days and human liver specimens from patients with primary biliary cholangitis (PBC) were collected to detect the expression of PGC1α. Hepatic-specific PGC1α knockout mice (HKO) were constructed and subjected to BDL, in which the effects of PGC1α on cholestatic liver injury were demonstrated by biochemical and histopathological assessments, immunoblotting, and metabolomics. RESULTS The expression of PGC1α was upregulated in the liver of PBC patients and murine models. Both in vivo and in vitro experiments supported the protective effects of PGC1α on cholestasis-induced hepatocyte injury. Infiltrated inflammatory cells after BDL were decreased in HKO mice. Inhibited Wnt/β-Catenin pathway and enhanced Notch signaling promoted transdifferentiation of hepatic progenitor cells (HPC)/ hepatocytes into cholangiocytes, leading to the greater ductular reaction observed in the HKO mice. But bile acids metabolism and mitochondrial function were not affected due to hepatic PGC1α deficiency in cholestasis. CONCLUSIONS Hepatic-specific deletion of PGC1α regulated liver regeneration by promoting ductular reactions, thereby exerting protective effects against BDL-induced liver injury, which could be a new potential therapeutic target.
Collapse
Affiliation(s)
- Dingwu Li
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Chenhui Ye
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Peihao Liu
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Ting Sun
- Department of Pathology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yunsheng Qin
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Xingyong Wan
- Department of Gastroenterology, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
32
|
Fadel L, Dacic M, Fonda V, Sokolsky BA, Quagliarini F, Rogatsky I, Uhlenhaut NH. Modulating glucocorticoid receptor actions in physiology and pathology: Insights from coregulators. Pharmacol Ther 2023; 251:108531. [PMID: 37717739 PMCID: PMC10841922 DOI: 10.1016/j.pharmthera.2023.108531] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Glucocorticoids (GCs) are a class of steroid hormones that regulate key physiological processes such as metabolism, immune function, and stress responses. The effects of GCs are mediated by the glucocorticoid receptor (GR), a ligand-dependent transcription factor that activates or represses the expression of hundreds to thousands of genes in a tissue- and physiological state-specific manner. The activity of GR is modulated by numerous coregulator proteins that interact with GR in response to different stimuli assembling into a multitude of DNA-protein complexes and facilitate the integration of these signals, helping GR to communicate with basal transcriptional machinery and chromatin. Here, we provide a brief overview of the physiological and molecular functions of GR, and discuss the roles of GR coregulators in the immune system, key metabolic tissues and the central nervous system. We also present an analysis of the GR interactome in different cells and tissues, which suggests tissue-specific utilization of GR coregulators, despite widespread functions shared by some of them.
Collapse
Affiliation(s)
- Lina Fadel
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Marija Dacic
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Physiology, Biophysics and Systems Biology, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Vlera Fonda
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Baila A Sokolsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
| | - Fabiana Quagliarini
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany
| | - Inez Rogatsky
- Hospital for Special Surgery Research Institute, The David Rosenzweig Genomics Center, New York, NY, USA; Graduate Program in Immunology and Microbial Pathogenesis, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA.
| | - N Henriette Uhlenhaut
- Institute for Diabetes and Endocrinology IDE, Helmholtz Munich, Ingolstaedter Landstr. 1, 857649 Neuherberg, Germany; Metabolic Programming, TUM School of Life Sciences & ZIEL Institute for Food and Health, Gregor11 Mendel-Str. 2, 85354 Freising, Germany.
| |
Collapse
|
33
|
Daniels LJ, Kay D, Marjot T, Hodson L, Ray DW. Circadian regulation of liver metabolism: experimental approaches in human, rodent, and cellular models. Am J Physiol Cell Physiol 2023; 325:C1158-C1177. [PMID: 37642240 PMCID: PMC10861179 DOI: 10.1152/ajpcell.00551.2022] [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: 12/19/2022] [Revised: 06/15/2023] [Accepted: 07/19/2023] [Indexed: 08/31/2023]
Abstract
Circadian rhythms are endogenous oscillations with approximately a 24-h period that allow organisms to anticipate the change between day and night. Disruptions that desynchronize or misalign circadian rhythms are associated with an increased risk of cardiometabolic disease. This review focuses on the liver circadian clock as relevant to the risk of developing metabolic diseases including nonalcoholic fatty liver disease (NAFLD), insulin resistance, and type 2 diabetes (T2D). Many liver functions exhibit rhythmicity. Approximately 40% of the hepatic transcriptome exhibits 24-h rhythms, along with rhythms in protein levels, posttranslational modification, and various metabolites. The liver circadian clock is critical for maintaining glucose and lipid homeostasis. Most of the attention in the metabolic field has been directed toward diet, exercise, and rather little to modifiable risks due to circadian misalignment or disruption. Therefore, the aim of this review is to systematically analyze the various approaches that study liver circadian pathways, targeting metabolic liver diseases, such as diabetes, nonalcoholic fatty liver disease, using human, rodent, and cell biology models.NEW & NOTEWORTHY Over the past decade, there has been an increased interest in understanding the intricate relationship between circadian rhythm and liver metabolism. In this review, we have systematically searched the literature to analyze the various experimental approaches utilizing human, rodent, and in vitro cellular approaches to dissect the link between liver circadian rhythms and metabolic disease.
Collapse
Affiliation(s)
- Lorna J Daniels
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Danielle Kay
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Thomas Marjot
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Leanne Hodson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| | - David W Ray
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- NIHR Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
- Kavli Centre for Nanoscience Discovery, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
34
|
Zou H, Yu J, Li Z, Liu Y, Wang T, Li T, Lv C, Zhang J. In vitro, in vivo, and in silico evaluation of the glucocorticoid receptor antagonist activity of 3,6-dibromocarbazole. Food Chem Toxicol 2023; 180:114048. [PMID: 37734465 DOI: 10.1016/j.fct.2023.114048] [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: 08/31/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023]
Abstract
3,6-Dibromocarbazole is a novel environmental contaminant which is currently detected in several environmental media worldwide. This work aims to investigate the anti-glucocorticoid potency and endocrine disrupting effects of 3,6-dibromocarbazole. In vitro experiments indicated that 3,6-dibromocarbazole possessed glucocorticoid receptor (GR) antagonistic activity and inhibited dexamethasone-induced GR nuclear translocation. 3,6-Dibromocarbazole reduced the expression levels of glucocorticoid responsive genes including glucose-6-phosphatase (G6Pase), phosphoenolpyruvate carboxykinase (PEPCK), fatty acid synthase (FAS), and tyrosine aminotransferase (TAT), and further disrupted the protein expression of two key enzymes PEPCK and FAS in gluconeogenesis. In vivo experiments showed that 3,6-dibromocarbazole induced abnormal development of zebrafish embryos and disrupted the major neurohormones involved in activation of hypothalamic-pituitary-adrenocortical (HPA) axis in zebrafish larvae. The results of molecular docking and molecular dynamics simulation contributed to explain the antagonistic effect of 3,6-dibromocarbazole. Taken together, this work identified 3,6-dibromocarbazole as a GR antagonist, which might exert endocrine disrupting effects by interfering the pathway of gluconeogenesis.
Collapse
Affiliation(s)
- Haoyang Zou
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Jia Yu
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China
| | - Zhuolin Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Yao Liu
- College of Food and Bioengineering, Qiqihar University, Qiqihar, 161006, China
| | - Tuoyi Wang
- College of Food and Bioengineering, Qiqihar University, Qiqihar, 161006, China
| | - Tiezhu Li
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Chengyu Lv
- Institute of Agro-food Technology, Jilin Academy of Agricultural Sciences, Changchun, 130033, China
| | - Jie Zhang
- College of Food Science and Engineering, Jilin University, Changchun, 130062, China.
| |
Collapse
|
35
|
Ganesan K, Xu C, Liu Q, Sui Y, Chen J. Unraveling the Role of Hepatic PGC1α in Breast Cancer Invasion: A New Target for Therapeutic Intervention? Cells 2023; 12:2311. [PMID: 37759533 PMCID: PMC10529029 DOI: 10.3390/cells12182311] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Breast cancer (BC) is the most common cancer among women worldwide and the main cause of cancer deaths in women. Metabolic components are key risk factors for the development of non-alcoholic fatty liver disease (NAFLD), which may promote BC. Studies have reported that increasing PGC1α levels increases mitochondrial biogenesis, thereby increasing cell proliferation and metastasis. Moreover, the PGC1α/ERRα axis is a crucial regulator of cellular metabolism in various tissues, including BC. However, it remains unclear whether NAFLD is closely associated with the risk of BC. Therefore, the present study aimed to determine whether hepatic PGC1α promotes BC cell invasion via ERRα. Various assays, including ELISA, western blotting, and immunoprecipitation, have been employed to explore these mechanisms. According to the KM plot and TCGA data, elevated PGC1α expression was highly associated with a shorter overall survival time in patients with BC. High concentrations of palmitic acid (PA) promoted PGC1α expression, lipogenesis, and inflammatory processes in hepatocytes. Conditioned medium obtained from PA-treated hepatocytes significantly increased BC cell proliferation. Similarly, recombinant PGC1α in E0771 and MCF7 cells promoted cell proliferation, migration, and invasion in vitro. However, silencing PGC1α in both BC cell lines resulted in a decrease in this trend. As determined by immunoprecipitation assay, PCG1a interacted with ERRα, thereby facilitating the proliferation of BC cells. This outcome recognizes the importance of further investigations in exploring the full potential of hepatic PGC1α as a prognostic marker for BC development.
Collapse
Affiliation(s)
- Kumar Ganesan
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Cong Xu
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Qingqing Liu
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Yue Sui
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
| | - Jianping Chen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Sassoon Road, Hong Kong, China; (K.G.); (C.X.); (Q.L.); (Y.S.)
- Shenzhen Institute of Research and Innovation, The University of Hong Kong, Shenzhen 518057, China
| |
Collapse
|
36
|
Mihaylov SR, Castelli LM, Lin YH, Gül A, Soni N, Hastings C, Flynn HR, Păun O, Dickman MJ, Snijders AP, Goldstone R, Bandmann O, Shelkovnikova TA, Mortiboys H, Ultanir SK, Hautbergue GM. The master energy homeostasis regulator PGC-1α exhibits an mRNA nuclear export function. Nat Commun 2023; 14:5496. [PMID: 37679383 PMCID: PMC10485026 DOI: 10.1038/s41467-023-41304-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
PGC-1α plays a central role in maintaining mitochondrial and energy metabolism homeostasis, linking external stimuli to transcriptional co-activation of genes involved in adaptive and age-related pathways. The carboxyl-terminus encodes a serine/arginine-rich (RS) region and an RNA recognition motif, however the RNA-processing function(s) were poorly investigated over the past 20 years. Here, we show that the RS domain of human PGC-1α directly interacts with RNA and the nuclear RNA export receptor NXF1. Inducible depletion of PGC-1α and expression of RNAi-resistant RS-deleted PGC-1α further demonstrate that its RNA/NXF1-binding activity is required for the nuclear export of some canonical mitochondrial-related mRNAs and mitochondrial homeostasis. Genome-wide investigations reveal that the nuclear export function is not strictly linked to promoter-binding, identifying in turn novel regulatory targets of PGC-1α in non-homologous end-joining and nucleocytoplasmic transport. These findings provide new directions to further elucidate the roles of PGC-1α in gene expression, metabolic disorders, aging and neurodegeneration.
Collapse
Affiliation(s)
- Simeon R Mihaylov
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
- Kinases and Brain Development Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Lydia M Castelli
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Ya-Hui Lin
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Aytac Gül
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Nikita Soni
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Christopher Hastings
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
| | - Helen R Flynn
- Proteomics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Oana Păun
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Mark J Dickman
- Department of Chemical and Biological Engineering, Sir Robert Hadfield Building, University of Sheffield, Mappin Street, Sheffield, S1 3JD, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Ambrosius P Snijders
- Proteomics Science Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Life Science Mass Spectrometry, Bruker Daltonics, Banner Lane, Coventry, CV4 9GH, UK
| | - Robert Goldstone
- Bioinformatics and Biostatistics Science and Technology Platform, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Oliver Bandmann
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Tatyana A Shelkovnikova
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Heather Mortiboys
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK
| | - Sila K Ultanir
- Kinases and Brain Development Laboratory, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Guillaume M Hautbergue
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, University of Sheffield, 385 Glossop Road, Sheffield, S10 2HQ, UK.
- Neuroscience Institute, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
- Healthy Lifespan Institute (HELSI), University of Sheffield, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
37
|
Gonzalez-Rellan MJ, Fernández U, Parracho T, Novoa E, Fondevila MF, da Silva Lima N, Ramos L, Rodríguez A, Serrano-Maciá M, Perez-Mejias G, Chantada-Vazquez P, Riobello C, Veyrat-Durebex C, Tovar S, Coppari R, Woodhoo A, Schwaninger M, Prevot V, Delgado TC, Lopez M, Diaz-Quintana A, Dieguez C, Guallar D, Frühbeck G, Diaz-Moreno I, Bravo SB, Martinez-Chantar ML, Nogueiras R. Neddylation of phosphoenolpyruvate carboxykinase 1 controls glucose metabolism. Cell Metab 2023; 35:1630-1645.e5. [PMID: 37541251 PMCID: PMC10487638 DOI: 10.1016/j.cmet.2023.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/08/2023] [Accepted: 07/10/2023] [Indexed: 08/06/2023]
Abstract
Neddylation is a post-translational mechanism that adds a ubiquitin-like protein, namely neural precursor cell expressed developmentally downregulated protein 8 (NEDD8). Here, we show that neddylation in mouse liver is modulated by nutrient availability. Inhibition of neddylation in mouse liver reduces gluconeogenic capacity and the hyperglycemic actions of counter-regulatory hormones. Furthermore, people with type 2 diabetes display elevated hepatic neddylation levels. Mechanistically, fasting or caloric restriction of mice leads to neddylation of phosphoenolpyruvate carboxykinase 1 (PCK1) at three lysine residues-K278, K342, and K387. We find that mutating the three PCK1 lysines that are neddylated reduces their gluconeogenic activity rate. Molecular dynamics simulations show that neddylation of PCK1 could re-position two loops surrounding the catalytic center into an open configuration, rendering the catalytic center more accessible. Our study reveals that neddylation of PCK1 provides a finely tuned mechanism of controlling glucose metabolism by linking whole nutrient availability to metabolic homeostasis.
Collapse
Affiliation(s)
- María J Gonzalez-Rellan
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Uxía Fernández
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Tamara Parracho
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Eva Novoa
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Marcos F Fondevila
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Natalia da Silva Lima
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Lucía Ramos
- Department of Biochemistry, CIMUS, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Amaia Rodríguez
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Endocrinology & Nutrition, Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, IdiSNA, Pamplona, Navarra, Spain
| | - Marina Serrano-Maciá
- Liver Disease Lab, BRTA CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Gonzalo Perez-Mejias
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla-CSIC. Avda. Americo Vespucio 49, 41092 Sevilla, Spain
| | - Pilar Chantada-Vazquez
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15705, A Coruña, Spain
| | - Cristina Riobello
- Gene Regulatory Control in Disease, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Christelle Veyrat-Durebex
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sulay Tovar
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Roberto Coppari
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Diabetes Center, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ashwin Woodhoo
- Gene Regulatory Control in Disease, CIMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Markus Schwaninger
- University of Lübeck, Institute for Experimental and Clinical Pharmacology and Toxicology, Lübeck, Germany
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, European Genomic Institute for Diabetes (EGID), 59000 Lille, France
| | - Teresa C Delgado
- Liver Disease Lab, BRTA CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain
| | - Miguel Lopez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Antonio Diaz-Quintana
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla-CSIC. Avda. Americo Vespucio 49, 41092 Sevilla, Spain
| | - Carlos Dieguez
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Diana Guallar
- Department of Biochemistry, CIMUS, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Gema Frühbeck
- CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Endocrinology & Nutrition, Metabolic Research Laboratory, Clínica Universidad de Navarra, University of Navarra, IdiSNA, Pamplona, Navarra, Spain
| | - Irene Diaz-Moreno
- Instituto de Investigaciones Químicas (IIQ), Centro de Investigaciones Científicas Isla de la Cartuja (cicCartuja), Universidad de Sevilla-CSIC. Avda. Americo Vespucio 49, 41092 Sevilla, Spain
| | - Susana B Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela 15705, A Coruña, Spain
| | - Maria L Martinez-Chantar
- Liver Disease Lab, BRTA CIC bioGUNE, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Derio, Bizkaia, Spain.
| | - Ruben Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; CIBER Fisiopatologia de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| |
Collapse
|
38
|
Abounouh K, Tanouti IA, Ouladlahsen A, Tahiri M, Badre W, Dehbi H, Sarih M, Benjelloun S, Pineau P, Ezzikouri S. The peroxisome proliferator-activated receptor γ coactivator-1 alpha rs8192678 (Gly482Ser) variant and hepatitis B virus clearance. Infect Dis (Lond) 2023; 55:614-624. [PMID: 37376899 DOI: 10.1080/23744235.2023.2228403] [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] [Received: 01/09/2023] [Revised: 06/06/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Chronic hepatitis B virus (CHB) infection is still incurable a major public health problem. It is yet unclear how host genetic factors influence the development of HBV infection. The peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) has been shown to regulate hepatitis B virus (HBV). Several reports found that PPARGC1A variants are involved in a number of distinct liver diseases. Here we investigate whether the PPARGC1A rs8192678 (Gly482Ser) variant is involved in the spontaneous clearance of acute HBV infection and if it participates in chronic disease progression in Moroccan patients. METHODS Our study included 292 chronic hepatitis B (CHB) patients and 181 individuals who spontaneously cleared-HBV infection. We genotyped the rs8192678 SNP using a TaqMan allelic discrimination assay and then explored its association with spontaneous HBV clearance and CHB progression. RESULTS Our data showed that individuals carrying CT and TT genotypes were more likely to achieve spontaneous clearance (OR = 0.48, 95% CI (0.32-0.73), p = 0.00047; OR = 0.28, 95% CI (0.15-0.53), p = 0.00005, respectively). Subjects carrying the mutant allele T were more likely to achieve spontaneous clearance (OR = 0.51, 95% CI (0.38-0.67), P = 2.68E-06). However, when we investigated the impact of rs8192678 on the progression of liver diseases, we neither observe any influence (p > 0.05) nor found any significant association between ALT, AST, HBV viral loads, and the PPARGC1A rs8192678 genotypes in patients with CHB (p > 0.05). CONCLUSION Our result suggests that PPARGC1A rs8192678 may modulate acute HBV infection, and could therefore represent a potential predictive marker in the Moroccan population.
Collapse
Affiliation(s)
- Karima Abounouh
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
- Laboratory of Cellular and Molecular Pathology, Medical School, University Hassan II
| | - Ikram-Allah Tanouti
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Ahd Ouladlahsen
- Faculté de médecine de Casablanca, CHU Ibn Rochd, Casablanca, Morocco
| | - Mohamed Tahiri
- Faculté de médecine de Casablanca, CHU Ibn Rochd, Casablanca, Morocco
| | - Wafaa Badre
- Faculté de médecine de Casablanca, CHU Ibn Rochd, Casablanca, Morocco
| | - Hind Dehbi
- Laboratory of Cellular and Molecular Pathology, Medical School, University Hassan II
| | - M'hammed Sarih
- Service de Parasitologie et des Maladies Vectorielles, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Soumaya Benjelloun
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Pascal Pineau
- Unité "Organisation Nucléaire et Oncogenèse", INSERM U993, Institut Pasteur, Paris, France
| | - Sayeh Ezzikouri
- Virology Unit, Viral Hepatitis Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| |
Collapse
|
39
|
Yang YN, Zhang MQ, Yu FL, Han B, Bao MY, Yan-He, Li X, Zhang Y. Peroxisom proliferator-activated receptor-γ coactivator-1α in neurodegenerative disorders: A promising therapeutic target. Biochem Pharmacol 2023; 215:115717. [PMID: 37516277 DOI: 10.1016/j.bcp.2023.115717] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/26/2023] [Accepted: 07/26/2023] [Indexed: 07/31/2023]
Abstract
Neurodegenerative disorders (NDDs) are characterized by progressive loss of selectively vulnerable neuronal populations and myelin sheath, leading to behavioral and cognitive dysfunction that adversely affect the quality of life. Identifying novel therapies that attenuate the progression of NDDs would be of significance. Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α), a widely expressed transcriptional regulator, modulates the expression of genes engaged in mitochondrial biosynthesis, metabolic regulation, and oxidative stress (OS). Emerging evidences point to the strong connection between PGC-1α and NDDs, suggesting its positive impaction on the progression of NDDs. Therefore, it is urgent to gain a deeper and broader understanding between PGC-1α and NDDs. To this end, this review presents a comprehensive overview of PGC-1α, including its basic characteristics, the post-translational modulations, as well as the interacting transcription factors. Secondly, the pathogenesis of PGC-1α in various NDDs, such as Alzheimer's (AD), Parkinson's (PD), and Huntington's disease (HD) is briefly discussed. Additionally, this study summarizes the underlying mechanisms that PGC-1α is neuroprotective in NDDs via regulating neuroinflammation, OS, and mitochondrial dysfunction. Finally, we briefly outline the shortcomings of current NDDs drug therapy, and summarize the functions and potential applications of currently available PGC-1α modulators (activator or inhibitors). Generally, this review updates our insight of the important role of PGC-1α on the development of NDDs, and provides a promising therapeutic target/ drug for the treatment of NDDs.
Collapse
Affiliation(s)
- Ya-Na Yang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Mao-Qing Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Feng-Lin Yu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Bing Han
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Ming-Yue Bao
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yan-He
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuan Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
40
|
Kumar A, Schwab M, Laborit Labrada B, Silveira MAD, Goudreault M, Fournier É, Bellmann K, Beauchemin N, Gingras AC, Bilodeau S, Laplante M, Marette A. SHP-1 phosphatase acts as a coactivator of PCK1 transcription to control gluconeogenesis. J Biol Chem 2023; 299:105164. [PMID: 37595871 PMCID: PMC10504565 DOI: 10.1016/j.jbc.2023.105164] [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: 12/02/2022] [Revised: 07/25/2023] [Accepted: 07/28/2023] [Indexed: 08/20/2023] Open
Abstract
We previously reported that the protein-tyrosine phosphatase SHP-1 (PTPN6) negatively regulates insulin signaling, but its impact on hepatic glucose metabolism and systemic glucose control remains poorly understood. Here, we use co-immunoprecipitation assays, chromatin immunoprecipitation sequencing, in silico methods, and gluconeogenesis assay, and found a new mechanism whereby SHP-1 acts as a coactivator for transcription of the phosphoenolpyruvate carboxykinase 1 (PCK1) gene to increase liver gluconeogenesis. SHP-1 is recruited to the regulatory regions of the PCK1 gene and interacts with RNA polymerase II. The recruitment of SHP-1 to chromatin is dependent on its association with the transcription factor signal transducer and activator of transcription 5 (STAT5). Loss of SHP-1 as well as STAT5 decrease RNA polymerase II recruitment to the PCK1 promoter and consequently PCK1 mRNA levels leading to blunted gluconeogenesis. This work highlights a novel nuclear role of SHP-1 as a key transcriptional regulator of hepatic gluconeogenesis adding a new mechanism to the repertoire of SHP-1 functions in metabolic control.
Collapse
Affiliation(s)
- Amit Kumar
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Michael Schwab
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Beisy Laborit Labrada
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Maruhen Amir Datsch Silveira
- Centre de Recherche du CHU de Québec - Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| | - Marilyn Goudreault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada; Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada
| | - Éric Fournier
- Centre de Recherche du CHU de Québec - Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada; Centre de recherche en données massives de l'Université Laval, Québec, Quebec, Canada
| | - Kerstin Bellmann
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada
| | - Nicole Beauchemin
- Department of Oncology, Medicine and Biochemistry, Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, Quebec, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, Ontario, Canada; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Steve Bilodeau
- Centre de Recherche du CHU de Québec - Université Laval, Axe Oncologie, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Département de biologie moléculaire, biochimie médicale et pathologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada; Centre de recherche en données massives de l'Université Laval, Québec, Quebec, Canada
| | - Mathieu Laplante
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada; Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada
| | - André Marette
- Faculté de Médecine, Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Université Laval, Québec, Quebec, Canada; Institute of Nutrition and Functional Foods, Laval University, Québec, Quebec, Canada.
| |
Collapse
|
41
|
Sacco SA, McAtee Pereira AG, Trenary I, Smith KD, Betenbaugh MJ, Young JD. Overexpression of peroxisome proliferator-activated receptor γ co-activator-1⍺ (PGC-1⍺) in Chinese hamster ovary cells increases oxidative metabolism and IgG productivity. Metab Eng 2023; 79:108-117. [PMID: 37473833 DOI: 10.1016/j.ymben.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 04/17/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
Chinese hamster ovary (CHO) cells are used extensively to produce protein therapeutics, such as monoclonal antibodies (mAbs), in the biopharmaceutical industry. MAbs are large proteins that are energetically demanding to synthesize and secrete; therefore, high-producing CHO cell lines that are engineered for maximum metabolic efficiency are needed to meet increasing demands for mAb production. Previous studies have identified that high-producing cell lines possess a distinct metabolic phenotype when compared to low-producing cell lines. In particular, it was found that high mAb production is correlated to lactate consumption and elevated TCA cycle flux. We hypothesized that enhancing flux through the mitochondrial TCA cycle and oxidative phosphorylation would lead to increased mAb productivities and final titers. To test this hypothesis, we overexpressed peroxisome proliferator-activated receptor γ co-activator-1⍺ (PGC-1⍺), a gene that promotes mitochondrial metabolism, in an IgG-producing parental CHO cell line. Stable cell pools overexpressing PGC-1⍺ exhibited increased oxygen consumption, indicating increased mitochondrial metabolism, as well as increased mAb specific productivity compared to the parental line. We also performed 13C metabolic flux analysis (MFA) to quantify how PGC-1⍺ overexpression alters intracellular metabolic fluxes, revealing not only increased TCA cycle flux, but global upregulation of cellular metabolic activity. This study demonstrates the potential of rationally engineering the metabolism of industrial cell lines to improve overall mAb productivity and to increase the abundance of high-producing clones in stable cell pools.
Collapse
Affiliation(s)
- Sarah A Sacco
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | | | - Irina Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Kevin D Smith
- Pharmaceutical Development and Manufacturing Sciences, Janssen Research and Development, Spring House, PA, USA
| | - Michael J Betenbaugh
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
42
|
Belosludtseva NV, Serov DA, Starinets VS, Penkov NV, Belosludtsev KN. Alterations in Mitochondrial Morphology and Quality Control in Primary Mouse Lung Microvascular Endothelial Cells and Human Dermal Fibroblasts under Hyperglycemic Conditions. Int J Mol Sci 2023; 24:12485. [PMID: 37569860 PMCID: PMC10419261 DOI: 10.3390/ijms241512485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/13/2023] Open
Abstract
The effect of hyperglycemia on the morphology of individual mitochondria and the state of the mitochondrial network in primary mouse lung microvascular endotheliocytes and human dermal fibroblasts has been investigated. The cells were exposed to high (30 mM) and low (5.5 mM) glucose concentrations for 36 h. In primary endotheliocytes, hyperglycemic stress induced a significant increase in the number of mitochondria and a decrease in the interconnectivity value of the mitochondrial network, which was associated with a decrease in the mean size of the mitochondria. Analysis of the mRNA level of the genes of proteins responsible for mitochondrial biogenesis and mitophagy revealed an increase in the expression level of the Ppargc1a, Pink1, and Parkin genes, indicating stimulated mitochondrial turnover in endotheliocytes under high glucose conditions. In primary fibroblasts, hyperglycemia caused a decrease in the number of mitochondria and an increase in their size. As a result, the mitochondria exhibited higher values for elongation. In parallel, the mRNA level of the Ppargc1a and Mfn2 genes in fibroblasts exposed to hyperglycemia was reduced. These findings indicate that high glucose concentrations induced cell-specific morphological rearrangements of individual mitochondria and the mitochondrial network, which may be relevant during mitochondria-targeted drug testing and therapy for hyperglycemic and diabetic conditions.
Collapse
Affiliation(s)
- Natalia V. Belosludtseva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Dmitriy A. Serov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, Vavilov St. 38, 119991 Moscow, Russia;
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Vlada S. Starinets
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Nikita V. Penkov
- Institute of Cell Biophysics, Russian Academy of Sciences, Institutskaya 3, 142290 Pushchino, Russia;
| | - Konstantin N. Belosludtsev
- Department of Biochemistry, Cell Biology and Microbiology, Mari State University, pl. Lenina 1, 424001 Yoshkar-Ola, Russia
| |
Collapse
|
43
|
Radi SH, Vemuri K, Martinez-Lomeli J, Sladek FM. HNF4α isoforms: the fraternal twin master regulators of liver function. Front Endocrinol (Lausanne) 2023; 14:1226173. [PMID: 37600688 PMCID: PMC10438950 DOI: 10.3389/fendo.2023.1226173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 07/18/2023] [Indexed: 08/22/2023] Open
Abstract
In the more than 30 years since the purification and cloning of Hepatocyte Nuclear Factor 4 (HNF4α), considerable insight into its role in liver function has been gleaned from its target genes and mouse experiments. HNF4α plays a key role in lipid and glucose metabolism and intersects with not just diabetes and circadian rhythms but also with liver cancer, although much remains to be elucidated about those interactions. Similarly, while we are beginning to elucidate the role of the isoforms expressed from its two promoters, we know little about the alternatively spliced variants in other portions of the protein and their impact on the 1000-plus HNF4α target genes. This review will address how HNF4α came to be called the master regulator of liver-specific gene expression with a focus on its role in basic metabolism, the contributions of the various isoforms and the intriguing intersection with the circadian clock.
Collapse
Affiliation(s)
- Sarah H. Radi
- Department of Biochemistry, University of California, Riverside, Riverside, CA, United States
| | - Kiranmayi Vemuri
- Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
- Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| | - Jose Martinez-Lomeli
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| | - Frances M. Sladek
- Department of Molecular, Cell and Systems Biology, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
44
|
Wu CC, Chen WC, Hsiao WP, Huang KF, Liao YS, Lin HB, Wu YJ, Kao CH, Chen SL. Reciprocal Regulation of Peroxisome Biogenesis and Myogenic Factors Is Critical for Myogenesis. Int J Mol Sci 2023; 24:12262. [PMID: 37569637 PMCID: PMC10419124 DOI: 10.3390/ijms241512262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/25/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Mitochondria (MITO) and peroxisomes (PEXO) are the major organelles involved in the oxidative metabolism of cells, but detailed examination of their dynamics and functional adaptations during skeletal muscle (SKM) development (myogenesis) is still lacking. In this study, we found that during myogenesis, MITO DNA, ROS level, and redox ratio increased in myotubes, but the membrane potential (Δψm) and ATP content reduced, implying that the MITO efficiency might reduce during myogenesis. The PEXO number and density both increased during myogenesis, which probably resulted from the accumulation and increased biogenesis of PEXO. The expression of PEXO biogenesis factors was induced during myogenesis in vitro and in utero, and their promoters were also activated by MyoD. Knockdown of the biogenesis factors Pex3 repressed not only the PEXO density and functions but also the levels of MITO genes and functions, suggesting a close coupling between PEXO biogenesis and MITO functions. Surprisingly, Pex3 knockdown by the CRISPRi system repressed myogenic differentiation, indicating critical involvement of PEXO biogenesis in myogenesis. Taken together, these observations suggest that the dynamics and functions of both MITO and PEXO are coupled with each other and with the metabolic changes that occur during myogenesis, and these metabolic couplings are critical to myogenesis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Shen-Liang Chen
- Department of Life Sciences, College of Health Sciences and Technology, National Central University, Taoyuan 320317, Taiwan; (C.-C.W.); (W.-C.C.); (W.-P.H.); (K.-F.H.); (Y.-S.L.); (H.-B.L.); (Y.-J.W.); (C.-H.K.)
| |
Collapse
|
45
|
Wang G, Qin S, Geng H, Zheng Y, Li R, Xia C, Chen L, Yao J, Deng L. Resveratrol Promotes Gluconeogenesis by Inhibiting SESN2-mTORC2-AKT Pathway in Calf Hepatocytes. J Nutr 2023; 153:1930-1943. [PMID: 37182694 DOI: 10.1016/j.tjnut.2023.05.005] [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: 03/02/2023] [Revised: 04/27/2023] [Accepted: 05/02/2023] [Indexed: 05/16/2023] Open
Abstract
BACKGROUND The glucose requirement of dairy cows is mainly met by increasing the rate of hepatic gluconeogenesis. However, due to negative energy balance, the liver of periparturient cows is under oxidative stress induced by lipid over-mobilization, and hepatic gluconeogenesis is reduced. Studies have demonstrated that resveratrol, which is widely known for its antioxidant properties, can alter hepatic gluconeogenesis. However, it is not clear whether resveratrol could regulate hepatic gluconeogenesis by its antioxidant properties. OBJECTIVES This study aims to investigate the precise effect of resveratrol in hepatic gluconeogenesis, the role of resveratrol on hydrogen peroxide (H2O2)-induced oxidative stress in hepatocytes and the potential mechanism using primary hepatocytes. METHODS Primary hepatocytes were isolated from 5 healthy Holstein calves (1 d old, 30 to 40 kg, fasted) and treated with different concentrations of resveratrol (0, 5, 10, 25, or 50 μM) combined with or without H2O2 (0, 100, or 200 μM) induction for 12 h. RESULTS Resveratrol enhanced the expression of gluconeogenic genes of calf hepatocytes in a dose-dependent manner (P < 0.05). Conversely, H2O2 suppressed the expression of gluconeogenic genes and induced oxidative stress (P < 0.05), which was improved by resveratrol in calf hepatocytes (P < 0.001). Furthermore, the mechanistic target of rapamycin complex 2 (mTORC2)-AKT pathway was found to negatively regulate gluconeogenesis. An AKT inhibitor was used to assess the role of the mTORC2-AKT pathway in the effects of resveratrol. The results showed resveratrol promoted hepatic gluconeogenesis by inhibiting the mTORC2-AKT pathway. Moreover, sestrin 2 (SESN2) upregulated the activity of mTORC2. We further found that resveratrol decreased SESN2 levels (P < 0.001). CONCLUSIONS This study indicated that resveratrol enhances the gluconeogenic capacity of calf hepatocytes by improving H2O2-induced oxidative stress and modulating the activity of the SESN2-mTORC2-AKT pathway, implying that resveratrol may be a promising target for ameliorating liver oxidative stress in transition cows.
Collapse
Affiliation(s)
- GuoYan Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - SenLin Qin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - HuiJun Geng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Yining Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Rongnuo Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Chao Xia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Lei Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - JunHu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China.
| |
Collapse
|
46
|
Rah SY, Joe Y, Park J, Ryter SW, Park C, Chung HT, Kim UH. CD38/ADP-ribose/TRPM2-mediated nuclear Ca 2+ signaling is essential for hepatic gluconeogenesis in fasting and diabetes. Exp Mol Med 2023; 55:1492-1505. [PMID: 37394593 PMCID: PMC10393965 DOI: 10.1038/s12276-023-01034-9] [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: 08/18/2022] [Revised: 02/06/2023] [Accepted: 04/17/2023] [Indexed: 07/04/2023] Open
Abstract
Hepatic glucose production by glucagon is crucial for glucose homeostasis during fasting, yet the underlying mechanisms remain incompletely delineated. Although CD38 has been detected in the nucleus, its function in this compartment is unknown. Here, we demonstrate that nuclear CD38 (nCD38) controls glucagon-induced gluconeogenesis in primary hepatocytes and liver in a manner distinct from CD38 occurring in the cytoplasm and lysosomal compartments. We found that the localization of CD38 in the nucleus is required for glucose production by glucagon and that nCD38 activation requires NAD+ supplied by PKCδ-phosphorylated connexin 43. In fasting and diabetes, nCD38 promotes sustained Ca2+ signals via transient receptor potential melastatin 2 (TRPM2) activation by ADP-ribose, which enhances the transcription of glucose-6 phosphatase and phosphoenolpyruvate carboxykinase 1. These findings shed light on the role of nCD38 in glucagon-induced gluconeogenesis and provide insight into nuclear Ca2+ signals that mediate the transcription of key genes in gluconeogenesis under physiological conditions.
Collapse
Affiliation(s)
- So-Young Rah
- Department of Biochemistry and National Creative Research Laboratory for Ca2+ Signaling Network, Jeonbuk National University, Medical School, Keum-am dong, Jeonju, 54907, Republic of Korea
| | - Yeonsoo Joe
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Jeongmin Park
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | | | - Chansu Park
- Department of Biochemistry and National Creative Research Laboratory for Ca2+ Signaling Network, Jeonbuk National University, Medical School, Keum-am dong, Jeonju, 54907, Republic of Korea
| | - Hun Taeg Chung
- School of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Uh-Hyun Kim
- Department of Biochemistry and National Creative Research Laboratory for Ca2+ Signaling Network, Jeonbuk National University, Medical School, Keum-am dong, Jeonju, 54907, Republic of Korea.
- Department of Biochemistry, School of Medicine, Wonkwang University, Iksan, 54538, Republic of Korea.
| |
Collapse
|
47
|
Uehara K, Santoleri D, Whitlock AEG, Titchenell PM. Insulin Regulation of Hepatic Lipid Homeostasis. Compr Physiol 2023; 13:4785-4809. [PMID: 37358513 PMCID: PMC10760932 DOI: 10.1002/cphy.c220015] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
The incidence of obesity, insulin resistance, and type II diabetes (T2DM) continues to rise worldwide. The liver is a central insulin-responsive metabolic organ that governs whole-body metabolic homeostasis. Therefore, defining the mechanisms underlying insulin action in the liver is essential to our understanding of the pathogenesis of insulin resistance. During periods of fasting, the liver catabolizes fatty acids and stored glycogen to meet the metabolic demands of the body. In postprandial conditions, insulin signals to the liver to store excess nutrients into triglycerides, cholesterol, and glycogen. In insulin-resistant states, such as T2DM, hepatic insulin signaling continues to promote lipid synthesis but fails to suppress glucose production, leading to hypertriglyceridemia and hyperglycemia. Insulin resistance is associated with the development of metabolic disorders such as cardiovascular and kidney disease, atherosclerosis, stroke, and cancer. Of note, nonalcoholic fatty liver disease (NAFLD), a spectrum of diseases encompassing fatty liver, inflammation, fibrosis, and cirrhosis, is linked to abnormalities in insulin-mediated lipid metabolism. Therefore, understanding the role of insulin signaling under normal and pathologic states may provide insights into preventative and therapeutic opportunities for the treatment of metabolic diseases. Here, we provide a review of the field of hepatic insulin signaling and lipid regulation, including providing historical context, detailed molecular mechanisms, and address gaps in our understanding of hepatic lipid regulation and the derangements under insulin-resistant conditions. © 2023 American Physiological Society. Compr Physiol 13:4785-4809, 2023.
Collapse
Affiliation(s)
- Kahealani Uehara
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dominic Santoleri
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna E. Garcia Whitlock
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Paul M. Titchenell
- Institute of Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
48
|
Deng X, Liang J, Wang L, Niu L, Xiao J, Guo Q, Liu X, Xiao C. Whole Grain Proso Millet ( Panicum miliaceum L.) Attenuates Hyperglycemia in Type 2 Diabetic Mice: Involvement of miRNA Profile. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37294881 DOI: 10.1021/acs.jafc.2c08184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
This work aimed to investigate the hypoglycemic effects and underlying mechanism of whole grain proso millet (Panicum miliaceum L.; WPM) on type 2 diabetes mellitus (T2DM). The results showed that WPM supplementation significantly reduced fasting blood glucose (FBG) and serum lipid levels in T2DM mice induced by a high-fat diet (HFD) combined with streptozotocin (STZ), with improved glucose tolerance, liver and kidney injury, and insulin resistance. In addition, WPM significantly inhibited the expression of gluconeogenesis-related genes G6pase, Pepck, Foxo1, and Pgc-1α. Further study by miRNA high-throughput sequencing revealed that WPM supplementation mainly altered the liver miRNA expression profile of T2DM mice by increasing the expression of miR-144-3p_R-1 and miR-423-5p, reducing the expression of miR-22-5p_R-1 and miR-30a-3p. GO and KEGG analyses showed that the target genes of these miRNAs were mainly enriched in the PI3K/AKT signaling pathway. WPM supplementation significantly increased the level of PI3K, p-AKT, and GSK3β in the liver of T2DM mice. Taken together, WPM exerts antidiabetic effects by improving the miRNA profile and activating the PI3K/AKT signaling pathway to inhibit gluconeogenesis. This study implies that PM can act as a dietary supplement to attenuate T2DM.
Collapse
Affiliation(s)
- Xu Deng
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Jiayi Liang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Lehui Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Li Niu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Jin Xiao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Qianqian Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Xuebo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| | - Chunxia Xiao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
49
|
Liu N, Yang X, Guo J, Zhang L, Huang S, Chen J, Huang J, Chen Y, Cui T, Zheng Y, Li T, Tang K, Zhong Y, Duan S, Yu L, Tang Y, Zheng D, Pan H, Gao Y. Hepatic ZBTB22 promotes hyperglycemia and insulin resistance via PEPCK1-driven gluconeogenesis. EMBO Rep 2023; 24:e56390. [PMID: 37154299 PMCID: PMC10240208 DOI: 10.15252/embr.202256390] [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] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 03/31/2023] [Accepted: 04/14/2023] [Indexed: 05/10/2023] Open
Abstract
Excessive gluconeogenesis can lead to hyperglycemia and diabetes through as yet incompletely understood mechanisms. Herein, we show that hepatic ZBTB22 expression is increased in both diabetic clinical samples and mice, being affected by nutritional status and hormones. Hepatic ZBTB22 overexpression increases the expression of gluconeogenic and lipogenic genes, heightening glucose output and lipids accumulation in mouse primary hepatocytes (MPHs), while ZBTB22 knockdown elicits opposite effects. Hepatic ZBTB22 overexpression induces glucose intolerance and insulin resistance, accompanied by moderate hepatosteatosis, while ZBTB22-deficient mice display improved energy expenditure, glucose tolerance, and insulin sensitivity, and reduced hepatic steatosis. Moreover, hepatic ZBTB22 knockout beneficially regulates gluconeogenic and lipogenic genes, thereby alleviating glucose intolerance, insulin resistance, and liver steatosis in db/db mice. ZBTB22 directly binds to the promoter region of PCK1 to enhance its expression and increase gluconeogenesis. PCK1 silencing markedly abolishes the effects of ZBTB22 overexpression on glucose and lipid metabolism in both MPHs and mice, along with the corresponding changes in gene expression. In conclusion, targeting hepatic ZBTB22/PEPCK1 provides a potential therapeutic approach for diabetes.
Collapse
Affiliation(s)
- Naihua Liu
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Key Specialty of Clinical PharmacyThe First Affiliated Hospital of Guangdong Pharmaceutical UniversityGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and ImmunologyXuzhou Medical UniversityXuzhouChina
| | - Jingyi Guo
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Lei Zhang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Shangyi Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jiabing Chen
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Jiawen Huang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yingjian Chen
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Tianqi Cui
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yi Zheng
- Faculty of Chinese MedicineMacau University of Science and TechnologyMacauChina
| | - Tianyao Li
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Kaijia Tang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Yadi Zhong
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Siwei Duan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
| | - Lili Yu
- Faculty of Chinese MedicineMacau University of Science and TechnologyMacauChina
| | - Ying Tang
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Dayong Zheng
- Department of HepatologyTCM‐Integrated Hospital of Southern Medical UniversityGuangzhouChina
- Department of HepatopancreatobiliaryCancer Center, Southern Medical UniversityGuangzhouChina
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese MedicineGuangzhou University of Chinese MedicineGuangzhouChina
| | - Huafeng Pan
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
| | - Yong Gao
- Science and Technology Innovation CenterGuangzhou University of Chinese MedicineGuangzhouChina
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine in Prevention and Treatment of TumorNanjing University of Chinese MedicineNanjingJiangsu ProvinceChina
- Division of Hypothalamic Research, Department of Internal MedicineThe University of Texas Southwestern Medical Center at DallasTXDallasUSA
| |
Collapse
|
50
|
Walls KM, Hong KU, Hein DW. Heterocyclic amines reduce insulin-induced AKT phosphorylation and induce gluconeogenic gene expression in human hepatocytes. Arch Toxicol 2023; 97:1613-1626. [PMID: 37005939 PMCID: PMC10192068 DOI: 10.1007/s00204-023-03488-2] [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: 09/23/2022] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
Heterocyclic amines (HCAs) are well-known for their mutagenic properties. One of the major routes of human exposure is through consumption of cooked meat, as certain cooking methods favor formation of HCAs. Recent epidemiological studies reported significant associations between dietary HCA exposure and insulin resistance and type II diabetes. However, no previous studies have examined if HCAs, independent of meat consumption, contributes to pathogenesis of insulin resistance or metabolic disease. In the present study, we have assessed the effect of three HCAs commonly found in cooked meat (2-amino-3,4,8-trimethylimidazo[4,5-f]quinoxaline [MeIQ], 2-amino-3,8-dimethylimidazo[4,5-f]quinoxaline [MeIQx], and 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine [PhIP]) on insulin signaling and glucose production. HepG2 or cryopreserved human hepatocytes were treated with 0-50 μM of MeIQ, MeIQx, or PhIP for 3 days. Treatment of HepG2 cells and hepatocytes with MeIQ and MeIQx resulted in a significant reduction in insulin-induced AKT phosphorylation, suggesting that HCA exposure decreases hepatic insulin signaling. HCA treatment also led to significant increases in expression of gluconeogenic genes, G6PC and PCK1, in both HepG2 and cryopreserved human hepatocytes. Additionally, the level of phosphorylated FOXO1, a transcriptional regulator of gluconeogenesis, was significantly reduced by HCA treatment in hepatocytes. Importantly, HCA treatment of human hepatocytes led to increases in extracellular glucose level in the presence of gluconeogenic substrates, suggesting that HCAs induce hepatic glucose production. The current findings suggest that HCAs induce insulin resistance and promote hepatic glucose production in human hepatocytes. This implicates that exposure to HCAs may lead to the development of type II diabetes or metabolic syndrome.
Collapse
Affiliation(s)
- Kennedy M. Walls
- Department of Pharmacology & Toxicology and Brown Cancer Center,
University of Louisville School of Medicine, Louisville, KY. U.S.A
| | - Kyung U. Hong
- Department of Pharmacology & Toxicology and Brown Cancer Center,
University of Louisville School of Medicine, Louisville, KY. U.S.A
| | - David W. Hein
- Department of Pharmacology & Toxicology and Brown Cancer Center,
University of Louisville School of Medicine, Louisville, KY. U.S.A
| |
Collapse
|