1
|
Jiang S, Liang Z, Hua J, Li Y, Fan X, Qiao Z, Wang Z, Shen Y, Fan L, Wang J. Network pharmacology and transcriptomics reveal Complanatoside A regulates lipid metabolism in hyperlipidemia and non-alcoholic fatty liver disease via the AMPK pathway. J Nutr Biochem 2025:109960. [PMID: 40354830 DOI: 10.1016/j.jnutbio.2025.109960] [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: 12/13/2024] [Revised: 04/28/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) and hyperlipidemia belong to the metabolic disorder syndromes of metabolic syndrome. They share a common pathological basis and are often complicated. Complanatoside A (CA), a flavonoid abundant in Astragali complanati semen, helps to prevent NAFLD and hyperlipidemia. However, the exact molecular mechanism is uncertain. Therefore, this study aims to explore the core mechanism. Network pharmacology was used to analyze the preventive mechanism of CA against NAFLD and hyperlipidemia. The efficacy of CA was proven in a high-fat diet-fed mouse model and a steatogenic hepatocyte model. Transcriptomic analysis, Western blot validation, and molecular docking methods were used to explore the common mechanism of CA in preventing NAFLD and hyperlipidemia. Network pharmacology revealed that the AMP-activated protein kinase (AMPK) pathway is a common mechanism leading to NAFLD and hyperlipidemia. It is also a potential pathway by which CA exerts its protective effect, which was confirmed in transcriptomics in vivo. Both in vitro and in vivo experiments showed that CA could inhibit lipid synthesis and promote fatty acid oxidation by activating the AMPK, alleviating lipid accumulation, and lipotoxic liver injury. This was demonstrated by the use of an AMPK inhibitor in vitro. Furthermore, molecular docking results showed that CA could directly interact with AMPK to regulate downstream lipid-related proteins. In conclusion, the AMPK pathway is key in developing NAFLD and hyperlipidemia. CA plays a dual preventive role in NAFLD and hyperlipidemia by activating AMPK to regulate lipid metabolism.
Collapse
Affiliation(s)
- Sijia Jiang
- Beijing University of Chinese Medicine, 102488, Beijing, China
| | | | - Jian Hua
- Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Yajin Li
- Xinjiang Medical University, 830011, Urumqi, China
| | - Xiaoxu Fan
- Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Zhiyuan Qiao
- Xinjiang Medical University, 830011, Urumqi, China
| | - Zhen Wang
- Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Yiwei Shen
- Beijing University of Chinese Medicine, 102488, Beijing, China
| | - Le Fan
- Department of Endocrinology, Xiyuan Hosipital of China Academy of Chinese Medical Sciences.
| | - Jingxia Wang
- Beijing University of Chinese Medicine, 102488, Beijing, China.
| |
Collapse
|
2
|
Tongtong P, Yujuan S, Ting L, Fangfang Y, Shijia W, Yilun H, Huadong Z, Qiongying Z, Yongping C, Dazhi C. Decoding ferroptosis in alcoholic hepatitis: A bioinformatics approach to hub gene identification. Genomics 2025; 117:111009. [PMID: 39864635 DOI: 10.1016/j.ygeno.2025.111009] [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: 08/27/2024] [Revised: 12/25/2024] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
BACKGROUND Ferroptosis is associated with alcoholic hepatitis (AH); however, the underlying mechanisms remain unclear. METHODS Changes in iron content and oxidative stress in AH patients and in vivo and in vitro models were analyzed. Iron homeostasis pathways in the livers of patients with AH were investigated using RNA sequencing. AH-associated ferroptosis-related genes (FRGs) were identified using weighted gene co-expression network analysis. Hub genes were identified using machine learning methods, and their diagnostic potential for AH was assessed. The correlation between FRGs and the immune microenvironment was analyzed, and the underlying regulatory mechanism was explored. FRG expression was validated in clinical samples and in vitro and in vivo models. The role of FRGs in AH-related ferroptosis was explored through gene-silencing experiments. RESULTS Significant iron deposits and oxidative stress were detected in clinical samples and in vivo and in vitro AH models. Bioinformatics identified GCLC, NQO1, and ULK1 as key FRGs linked to the immune microenvironment and AH-related pathogenic genes. A nomogram based on these FRGs accurately assessed AH risk, as validated using the calibration curve. A regulatory network involving 154 miRNAs and 136 transcription factors was mapped for FRGs. In AH patients, NQO1 was upregulated in the liver, whereas GCLC and ULK1 were downregulated. Silencing GCLC and ULK1 reduced cell viability and increased oxidative stress and ferroptosis, whereas silencing NQO1 had the opposite effect. CONCLUSIONS Therefore, GCLC, NQO1, and ULK1 are key AH-related FRGs, potentially serving as targets for diagnosing and treating AH.
Collapse
Affiliation(s)
- Pan Tongtong
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Shen Yujuan
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Huzhou Key Laboratory of Precision Medicine Research and Translation for Infectious Diseases, Huzhou Central Hospital, Wuxing District, Huzhou 313000, China
| | - Li Ting
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yi Fangfang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Wu Shijia
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Huang Yilun
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhang Huadong
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Zhang Qiongying
- Department of Pathology, the First Affiliated Hospital of Wenzhou Medical University, Ouhai District, Wenzhou 325035, China
| | - Chen Yongping
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Chen Dazhi
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; Hangzhou Medical College, Linan District, Hangzhou 311300, China.
| |
Collapse
|
3
|
Li X, Rao Z, Hu W, Lu W, Luo Y. Treating metabolic dysfunction-associated steatohepatitis: The fat-trimming FGF21 approach. Obes Rev 2025; 26:e13861. [PMID: 39546893 DOI: 10.1111/obr.13861] [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: 06/02/2023] [Revised: 09/10/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a condition characterized by hepatosteatosis, inflammation, and tissue damage, with steatosis as the initial stage, which involves chronic, excess deposition of lipids in hepatic lipid droplets. Despite the growing prevalence and serious risks it poses, including liver decompensation, the need for transplantation, and increased patient mortality, MASH currently faces no approved pharmacotherapy. Several promising treatment candidates have emerged from recent clinical trials, including analogs of FGF21 and agonists of the associated FGFR1-KLB complex. These agents were well-tolerated in trials and have demonstrated significant improvements in both histological and biochemical markers of liver fat content, inflammation, injury, and fibrosis in patients with MASH. Endocrine FGF21 plays a vital role in maintaining homeostasis of lipid, glucose, and energy metabolism. It achieves this through pathways that target lipids or lipid droplets in adipocytes and hepatocytes. Mechanistically, pharmacological FGF21 acts as a potent catabolic factor to promote lipid or lipid droplet lipolysis, fatty acid oxidation, mitochondrial catabolic flux, and heat-dissipating energy expenditure, leading to effective clearance of hepatic and systemic gluco-lipotoxicity and inflammatory stress, thereby preventing obesity, diabetes, and MASH pathologies. In this review, we aim to provide an update on the outcomes of clinical trials for several FGF21 mimetics. We compare these outcomes with preclinical studies and offer a lipid-centric perspective on the mechanisms underlying the clinical benefits of these agents for MASH.
Collapse
Affiliation(s)
- Xiaokun Li
- School of Pharmaceutical Sciences, Wenzhou Medical University, & Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, China
| | - Zhiheng Rao
- School of Pharmaceutical Sciences, Wenzhou Medical University, & Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, China
| | - Wenhao Hu
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiqin Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, El Paso, Texas, USA
| | - Yongde Luo
- School of Pharmaceutical Sciences, Wenzhou Medical University, & Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), Wenzhou, Zhejiang, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
4
|
Liang M, Xiao X, Chen M, Guo Y, Han W, Min Y, Jiang X, Yu W. Artemisia capillaris Thunb. Water extract alleviates metabolic dysfunction-associated Steatotic liver disease Disease by inhibiting miR-34a-5p to activate Sirt1-mediated hepatic lipid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119030. [PMID: 39515682 DOI: 10.1016/j.jep.2024.119030] [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: 08/03/2024] [Revised: 10/14/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia capillaris Thunb. (ACT) is a plant in the Asteraceae family. Its traditional effects are to clear away dampness and heat, promote gallbladder and reduce jaundice. Traditional Chinese medicine believes that MASLD is a damp-heat syndrome. The group's previous study showed that Artemisia capillaris Thunb. Water Extract (ACTE) has an improved effect on MASLD. AIM OF THE STUDY AND METHODS In order to further understand its mechanism of action, this study established a mouse MASLD model and a HepG2 cell lipid droplet model, combined small RNA sequencing and miRNA transfection experiments, to explore the mechanism of ACTE to improve MASLD by modulating miRNA-targeted mRNA. Non-targeted metabolomics method was used to detect and analyze ACTE. RESULTS This study screened miR-34a-5p and confirmed its target mRNA-Sirtuin 1 (Sirt1). MASLD induced high expression of miR-34a-5p and low expression of Sirt1, and ACE reversed these changes. When overexpressing miR-34a-5p or knocking down Sirt1, the effect of ACE in reducing PO (palmitic acid and oleic acid complex)-induced lipid accumulation in HepG2 cells was attenuated. ACTE reduces the expression of FASN, SCD1, ACC, and SREBP-1c, promotes the expression of CPT-1 and HSL, thereby reducing lipid accumulation. CONCLUSIONS ACTE activates Sirt1 by inhibiting the expression of miR-34a-5p, thereby reducing liver lipid accumulation and improving HFD-induced MASLD. These findings highlight the potential of ACTE in reducing weight, controlling obesity, and improving lipid metabolism disorders.
Collapse
Affiliation(s)
- Meng Liang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Xiao Xiao
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Miao Chen
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Yi Guo
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Weiting Han
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Yahong Min
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Xiaowen Jiang
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China.
| | - Wenhui Yu
- Department of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Institute of Chinese Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, China; Heilongjiang Provincial Key Laboratory of Animal Pathogenesis and Comparative Medicine, Harbin, 150030, China.
| |
Collapse
|
5
|
Xin W, Zhou J, Peng Y, Gong S, Liao W, Wang Y, Huang X, Mao Y, Yao M, Qin S, Xiong J, Li Y, Lan Q, Huang Y, Zhao J. SREBP1c-Mediated Transcriptional Repression of YME1L1 Contributes to Acute Kidney Injury by Inducing Mitochondrial Dysfunction in Tubular Epithelial Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412233. [PMID: 39680752 PMCID: PMC11809329 DOI: 10.1002/advs.202412233] [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: 10/01/2024] [Revised: 11/29/2024] [Indexed: 12/18/2024]
Abstract
Acute kidney injury (AKI) is a prevalent clinical syndrome with high morbidity and mortality. Accumulating studies suggest mitochondrial dysfunction as the typical characteristics and key process of AKI, but the underlying mechanism remains elusive. The YME1-like 1 (YME1L1) ATPase, an inner mitochondrial membrane protein, is screened and identified to be downregulated in renal tubular epithelial cells of various mouse models and patients of AKI. Dramatically, restoration of YME1L1 expression significantly alleviates cisplatin-induced AKI and subsequent chronic kidney disease (CKD) through attenuating mitochondrial dysfunction via maintaining optic atrophy 1 (OPA1)-mediated mitochondrial energy metabolism homeostasis. Mechanistically, the upregulated expression of sterol regulatory element binding transcription factor 1c (SREBP1c) is demonstrated to be responsible for cisplatin-mediated transcriptional inhibition of YME1L1 via directly binding to its promoter region. Moreover, cisplatin-induced methyltransferase-like 3 (METTL3)-mediated m6A modification enhances SREBP1c mRNA stability, thereby upregulating its expression. Notably, both depletion of SREBP1c and renal tubule-specific overexpression of YME1L1 markedly ameliorate cisplatin-induced AKI and its transition to CKD. Taken together, these findings suggest that METTL3-mediated SREBP1c upregulation contributes to AKI and its progression to CKD through disrupting mitochondrial energy metabolism via transcriptionally suppressing YME1L1. Targeting the SREBP1c/YME1L1 signaling may serve as a novel therapeutic strategy against AKI.
Collapse
Affiliation(s)
- Wang Xin
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jie Zhou
- Department of OncologySouthwest Cancer CenterSouthwest HospitalArmy Medical UniversityChongqing400038China
| | - Yuzhu Peng
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Shuiqin Gong
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Wenhao Liao
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yaqin Wang
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Xixin Huang
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yang Mao
- Clinical Medical Research CenterXinqiao HospitalArmy Medical UniversityChongqing400037China
| | - Mengying Yao
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Shaozong Qin
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jiachuan Xiong
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yan Li
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Qigang Lan
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Yinghui Huang
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| | - Jinghong Zhao
- Department of NephrologyChongqing Key Laboratory of Prevention and Treatment of Kidney DiseaseChongqing Clinical Research Center of Kidney and Urology DiseasesXinqiao HospitalArmy Medical University (Third Military Medical University)Chongqing400037China
| |
Collapse
|
6
|
Kolapalli SP, Beese CJ, Reid SE, Brynjólfsdóttir SH, Jørgensen MH, Jain A, Cuenco J, Lewinska M, Abdul-Al A, López AR, Jäättelä M, Sakamoto K, Andersen JB, Maeda K, Rusten TE, Lund AH, Frankel LB. Pellino 3 E3 ligase promotes starvation-induced autophagy to prevent hepatic steatosis. SCIENCE ADVANCES 2025; 11:eadr2450. [PMID: 39823344 PMCID: PMC11740972 DOI: 10.1126/sciadv.adr2450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 12/18/2024] [Indexed: 01/30/2025]
Abstract
Nutrient deprivation is a major trigger of autophagy, a conserved quality control and recycling process essential for cellular and tissue homeostasis. In a high-content image-based screen of the human ubiquitome, we here identify the E3 ligase Pellino 3 (PELI3) as a crucial regulator of starvation-induced autophagy. Mechanistically, PELI3 localizes to autophagic membranes, where it interacts with the ATG8 proteins through an LC3-interacting region (LIR). This facilitates PELI3-mediated ubiquitination of ULK1, driving ULK1's subsequent proteasomal degradation. PELI3 depletion leads to an aberrant accumulation and mislocalization of ULK1 and disrupts the early steps of autophagosome formation. Genetic deletion of Peli3 in mice impairs fasting-induced autophagy in the liver and enhances starvation-induced hepatic steatosis by reducing autophagy-mediated clearance of lipid droplets. Notably, PELI3 expression is decreased in the livers of patients with metabolic dysfunction-associated steatotic liver disease (MASLD), suggesting its role in hepatic steatosis development in humans. The findings suggest that PELI3-mediated control of autophagy plays a protective role in liver health.
Collapse
Affiliation(s)
- Srinivasa P. Kolapalli
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Carsten J. Beese
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Steven E. Reid
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | | | - Maria H. Jørgensen
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Ashish Jain
- Center for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Joyceline Cuenco
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Monika Lewinska
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
- Gubra, DK-2970 Hørsholm, Denmark
| | - Ahmad Abdul-Al
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Aida R. López
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Marja Jäättelä
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kenji Maeda
- Cell Death and Metabolism, Center for Autophagy, Recycling and Disease, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
| | - Tor E. Rusten
- Center for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Anders H. Lund
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Lisa B. Frankel
- Cellular Homeostasis and Recycling, Danish Cancer Institute, DK-2100 Copenhagen, Denmark
- Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
7
|
Amer AE, Ghoneim HA, Abdelaziz RR, Shehatou GSG, Suddek GM. L-carnitine attenuates autophagic flux, apoptosis, and necroptosis in rats with dexamethasone-induced non-alcoholic steatohepatitis. BMC Pharmacol Toxicol 2024; 25:102. [PMID: 39736705 DOI: 10.1186/s40360-024-00820-z] [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: 01/16/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
BACKGROUND UpToDate, no drugs have been approved to treat nonalcoholic steatohepatitis, the advanced stage of the most prevalent liver disease, non-alcoholic fatty liver disease. The present study was conducted to explore the potential influences of L-carnitine on the pathomechanisms of hepatic injury that mediate progression to non-alcoholic steatohepatitis in dexamethasone-toxified rats. METHODS Male Wistar rats were allocated as follows: dexamethasone group, rats received dexamethasone (8 mg/kg/day, intraperitoneally) for 6 days; DEXA-LCAR300, DEXA-LCAR500, and DEXA-MET groups, rats administered L-carnitine (300 or 500 mg/kg/day, IP) or metformin (500 mg/kg/day, orally) one week prior to dexamethasone injection (8 mg/kg/day, IP) and other six days alongside dexamethasone administration. Two groups of age-matched normal rats received either the drug vehicle (the control group) or the higher dose of L-carnitine (the drug-control group). At the end of the experiment, sets of biochemical, histological, and immunohistochemical examinations were performed. RESULTS L-carnitine (mainly at the dose of 500 mg/kg/day) markedly abolished dexamethasone-induced alterations in glucose tolerance, hepatic histological features, and serum parameters of hepatic function and lipid profile. Moreover, it significantly ameliorated dexamethasone-induced elevations of hepatic oxidative stress, SREBP-1 and p-MLKL protein levels, and nuclear FOXO1, LC3, P62, and caspase-3 immunohistochemical expression. Furthermore, it markedly diminished dexamethasone-induced suppression of hepatic Akt phosphorylation and Bcl2 immunohistochemical expression. The effects of L-carnitine (500 mg/kg/day) were comparable to those of metformin in most assessments and better than its corresponding lower dose. CONCLUSION These findings introduce L-carnitine as a potential protective drug that may mitigate the rate of disease progression in non-alcoholic fatty liver disease patients with early stages or those at the highest risks.
Collapse
Affiliation(s)
- Ahmed E Amer
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, Dakahliya, 35712, Egypt.
| | - Hamdy A Ghoneim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Rania R Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - George S G Shehatou
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, Dakahliya, 35712, Egypt
| | - Ghada M Suddek
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
8
|
Lu Q, La M, Wang Z, Huang J, Zhu J, Zhang D. Investigation of Active Components of Meconopsis integrifolia (Maxim.) Franch in Mitigating Non-Alcoholic Fatty Liver Disease. Int J Mol Sci 2024; 26:50. [PMID: 39795910 PMCID: PMC11719989 DOI: 10.3390/ijms26010050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has rapidly emerged as the most prevalent chronic liver disease globally, representing a significant and escalating public health challenge. Meconopsis integrifolia (Maxim.) Franch, a traditional Tibetan medicinal herb used for treating hepatitis, remains largely unexplored regarding its therapeutic potential and active components in combating NAFLD. This study first evaluated the in vitro lipid accumulation inhibitory activity of different extraction fractions of M. integrifolia using a HepG2 cell steatosis model. The ethyl acetate fraction was found to significantly reduce triglyceride (TG) and low-density lipoprotein (LDL) levels, inhibit lipid droplet deposition in HepG2 cells, and promote lipid metabolism balance through modulation of the AMPK/SREPB-1c/PPAR-α signaling pathway. Further analysis utilizing chromatographic techniques and nuclear magnetic resonance spectroscopy (NMR) led to the isolation of 13 compounds from the active ethyl acetate fraction. Notably, compounds 6, 9, 10, 11, 12, and 13 were identified for the first time from this Tibetan herb. In vitro activity assays and molecular docking analyses further confirmed that the compounds Luteolin (1), Quercetin 3-O-[2‴, 6‴-O-diacetyl-β-d-glucopyranosyl-(1→6)-β-d-glucopyranoside] (6), and Quercetin 3-O-[2‴-O-acetyl-β-d-glucopyranosyl-(1→6)-β-d-glucopyranoside] (8) are potential key components responsible for the NAFLD-ameliorating effects of M. integrifolia. This study highlights the therapeutic potential of M. integrifolia in treating NAFLD and provides a foundation for its further development and application, underscoring its significance in the advanced utilization of traditional Tibetan medicine.
Collapse
Affiliation(s)
- Qiqin Lu
- Research Center for High Altitude Medicine, Key Laboratory of the Ministry of High Altitude Medicine, Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China;
- College of Chemical Engineering, Qinghai University, Xining 810016, China; (M.L.); (Z.W.); (J.H.); (J.Z.)
| | - Majia La
- College of Chemical Engineering, Qinghai University, Xining 810016, China; (M.L.); (Z.W.); (J.H.); (J.Z.)
| | - Ziyang Wang
- College of Chemical Engineering, Qinghai University, Xining 810016, China; (M.L.); (Z.W.); (J.H.); (J.Z.)
| | - Jiaomei Huang
- College of Chemical Engineering, Qinghai University, Xining 810016, China; (M.L.); (Z.W.); (J.H.); (J.Z.)
| | - Jiahui Zhu
- College of Chemical Engineering, Qinghai University, Xining 810016, China; (M.L.); (Z.W.); (J.H.); (J.Z.)
| | - Dejun Zhang
- Research Center for High Altitude Medicine, Key Laboratory of the Ministry of High Altitude Medicine, Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China;
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China
| |
Collapse
|
9
|
Dong C, Huoshen W, Bai Y, Liu J, Li B, Guan Y, Luo P. Uncovering the molecular networks of ferroptosis in the pathogenesis of type 2 diabetes and its complications: a multi-omics investigation. Mol Med 2024; 30:268. [PMID: 39716081 DOI: 10.1186/s10020-024-01045-w] [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: 10/25/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Diabetes is a multi-factorial disorder and related complications constitute one of the principal causes of global mortality and disability. The role of ferroptosis in diabetes and its complications is intricate and significant. This study endeavors to disclose the role of ferroptosis in the aforementioned diseases from multiple perspectives through multi-omics. METHODS We performed genetic correlation analyses via the Linkage Disequilibrium Score and High-Definition Likelihood approaches for type 2 diabetes (T2D) and its complications. The data concerning the expression of ferroptosis-related genes (FRGs) were obtained from the meta-analysis of studies on gene expression and protein abundance. Mendelian randomization analyses and cross-validation were implemented using the discovery cohort, replication cohort, and imaging genomics cohort of T2D and its complications. Moreover, we conducted colocalization analyses on T2D and tissue-specific single-cell RNA sequencing investigations on the complications to complement the results. RESULTS Genetic association analysis indicated that the selected datasets could be incorporated into a secondary analysis of T2D complications. In the primary analysis, six FRGs (CDKN1A, ENO3, FURIN, RARRES2, TYRO3, and YTHDC2) were found to be positively associated with T2D risk. Conversely, eight FRGs (ARNTL, CAMKK2, CTSB, FADS2, KDM5A, MEG3, SREBF1, and STAT3) were inversely associated with T2D risk. The 14 FRGs were included in the secondary analysis. Within the FRGs, which received full support from both the discovery and replication cohorts, and were further validated by imaging genomics, higher levels of CDKN1A were positively associated with DKD risk. Higher levels of CAMKK2 and KDM5A were associated with a decreased risk of DKD. For DCM, higher levels of CTSB were positively associated with DCM risk. And genetically predicted higher levels of ARNTL and SREBF1 were associated with a decreased risk of NAFLD. Finally, we validated the tissue-specific expression of each complication with scRNA-seq datasets. CONCLUSIONS This study identified FRGs in relation to T2D and its complications, which may enhance the understanding of the pathogenic mechanisms of their development. Meanwhile, it offers cross-validation for imaging genomics and further indicates the direction for non-invasive diagnosis.
Collapse
Affiliation(s)
- Changqing Dong
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China
| | - Wuda Huoshen
- School of Stomatology, Southwest Medical University, Luzhou, Sichuan, China
| | - Yunfeng Bai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| | - Jiaona Liu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, National Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases Research, Beijing, China
| | - Bing Li
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China
| | - Yucan Guan
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China
| | - Ping Luo
- Department of Nephrology, National Key Laboratory of Diabetes, The Second Hospital of Jilin University, No. 991 Yatai Street, Nanguan District, Changchun, Jilin, China.
| |
Collapse
|
10
|
Shan B, Zhao C, Peng C, Miao Y, Lei S, Zhao L, Jia M, Pan S, Gong J, Wang Q. Theabrownin from Pu-erh tea attenuated high-fat diet-induced metabolic syndrome in rat by regulating microRNA and affecting gut microbiota. Int J Biol Macromol 2024; 285:138368. [PMID: 39638201 DOI: 10.1016/j.ijbiomac.2024.138368] [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: 08/28/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Theabrownin (TB), the primary pigment in Pu-erh tea, has shown potential in alleviating metabolic syndrome (MS), though its precise mechanisms remain unclear. This study investigated the effects of Pu-erh tea water extract (WE) and TB on high-fat diet (HFD)-induced MS in rats, focusing on miRNA regulation and gut microbiota composition. Both WE and TB significantly improved markers of MS, including dyslipidemia, insulin resistance, and inflammation. These improvements were linked to the normalization of specific miRNAs (miR-125b-5p, miR-223-3p_R + 2, miR-148b-3p, and miR-1247-5p), which activated the PI3K/AKT/FOXO signaling pathway, subsequently modulating key genes involved in glucolipid metabolism (SREBP-1C, PEPCK, PGC-1α, and G6pc). Additionally, WE and TB restored gut microbiota balance by decreasing the Firmicutes/Bacteroidetes ratio and increasing beneficial bacteria such as Bacteroides, Lactobacillus, and Bifidobacterium, while reducing harmful bacteria like Pseudomonas. These findings underscore the potential of theabrownin as a functional food component for MS prevention, offering new insights into its miRNA-mediated and microbiota-related mechanisms.
Collapse
Affiliation(s)
- Bo Shan
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chunyan Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Chunxiu Peng
- College of Horticulture and Landscape, Yunnan Agricultural University, Kunming 650201, China
| | - Yue Miao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Shuwen Lei
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Lei Zhao
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; College of Science, Yunnan Agricultural University, Kunming 650201, China.
| | - Man Jia
- Fermentation Engineering Research Center for Yunnan Pu-erh Tea, Kunming 650217, China
| | - Shukang Pan
- Fermentation Engineering Research Center for Yunnan Pu-erh Tea, Kunming 650217, China.
| | - Jiashun Gong
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Agro-Products Processing Research Institute, Yunnan Academy of Agricultural Sciences, Kunming 650223, China.
| | - Qiuping Wang
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
11
|
Zhu X, Cao Y, Chen S, Liu Q, Chai J, Wang W. Insufficient S-sulfhydration of serum and glucocorticoid-regulated kinase 1 participates in hyperhomocysteinemia-induced liver injury. Free Radic Biol Med 2024; 225:517-527. [PMID: 39427745 DOI: 10.1016/j.freeradbiomed.2024.10.294] [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: 09/05/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND & AIMS Previous studies have established that hyperhomocysteinemia (HHcy) significantly contributes to the development of non-alcoholic steatohepatitis (NASH). Conversely, hydrogen sulfide (H2S) has shown potential in mitigating NASH. Despite these findings, it remains uncertain whether H2S can serve as a therapeutic agent against HHcy-induced liver damage. METHODS Mice were fed a high-methionine diet to induce HHcy and HepG2 cells were exposed to homocysteine (Hcy). In both models, we assessed liver injury, H2S concentration, and autophagy levels. For rescue, sodium hydrosulfide (NaHS), an H2S donor, was used to test its potential in reversing hepatic pathological features induced by HHcy. RESULTS 1) Hcy accumulation led to liver damage and increased autophagy. This was linked to insufficient S-sulfhydration of serum and glucocorticoid-regulated kinase 1 (SGK1) at Cys244 and Cys282, a crucial autophagy regulator. The deficiency in S-sulfhydration was resulted from downregulation of cystathionine-γ-lyase (CSE) and subsequent H2S decrease, leading to SGK1 inactivation. 2) Administration of NaHS reduced the liver damage caused by high Hcy levels and restored H2S levels, promoting the S-sulfhydration and activation of SGK1. 3) Pharmacological inhibition of SGK1 induced autosis, a specific type of cell death caused by overactivation of autophagy. Conversely, a constitutively active mutant of SGK1 (SGK1S422D) significantly decreased autophagy and improved cell viability. CONCLUSIONS NaHS supplementation mitigates HHcy-induced liver injury by downregulating hepatic autophagy through the S-sulfhydration and activation of SGK1. This post-translational modification by H2S holds promise as a therapeutic approach for HHcy-induced liver injury.
Collapse
Affiliation(s)
- Xinyu Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yan Cao
- Department of Basic Medical Sciences, Beijing Health Vocational College, Beijing, 101149, China
| | - Shuai Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qinchi Liu
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Jiayin Chai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory for Metabolic Disorder-Related Cardiovascular Diseases, Beijing, 100069, China.
| |
Collapse
|
12
|
Marchiori GN, Eynard AR, Soria EA. Essential Fatty Acids along the Women’s Life Cycle and Promotion of a
Well-balanced Metabolism. CURRENT WOMENS HEALTH REVIEWS 2024; 20. [DOI: 10.2174/0115734048247312230929092327] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 07/12/2023] [Accepted: 08/21/2023] [Indexed: 01/03/2025]
Abstract
Abstract:
Linoleic acid (ω-6 LA) and α-linolenic acid (ω-3 ALA) are essential fatty acids (EFA)
for human beings. They must be consumed through diet and then extensively metabolized, a process that plays a fundamental role in health and eventually in disease prevention. Given the numerous changes depending on age and sex, EFA metabolic adaptations require further investigations
along the women’s life cycle, from onset to decline of the reproductive age. Thus, this review explains women’s life cycle stages and their involvement in diet intake, digestion and absorption,
the role of microbiota, metabolism, bioavailability, and EFA fate and major metabolites. This
knowledge is crucial to promoting lipid homeostasis according to female physiology through well-directed health strategies. Concerning this, the promotion of breastfeeding, nutrition, and physical activity is cardinal to counteract ALA deficiency, LA/ALA imbalance, and the release of unhealthy derivatives. These perturbations arise after menopause that compromise both lipogenic
and lipolytic pathways. The close interplay of diet, age, female organism, and microbiota also
plays a central role in regulating lipid metabolism. Consequently, future studies are encouraged to
propose efficient interventions for each stage of women's cycle. In this sense, plant-derived foods
and products are promising to be included in women’s nutrition to improve EFA metabolism.
Collapse
Affiliation(s)
- Georgina N. Marchiori
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Cátedra de Biología Celular, Histología y Embriología,
Instituto de Biología Celular. Bv. de la Reforma, Ciudad Universitaria, 5014, Córdoba, Argentina
- Universidad
Nacional de Córdoba, Facultad de Ciencias Médicas, Escuela de Nutrición. Bv. de la Reforma, Ciudad Universitaria,
5014, Córdoba, Argentina
| | - Aldo R. Eynard
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Cátedra de Biología Celular, Histología y Embriología,
Instituto de Biología Celular. Bv. de la Reforma, Ciudad Universitaria, 5014, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, INICSA.
Bv. de la Reforma, Ciudad Universitaria, 5014, Córdoba, Argentina
| | - Elio A. Soria
- Universidad Nacional de Córdoba, Facultad de Ciencias Médicas, Cátedra de Biología Celular, Histología y Embriología,
Instituto de Biología Celular. Bv. de la Reforma, Ciudad Universitaria, 5014, Córdoba, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, INICSA.
Bv. de la Reforma, Ciudad Universitaria, 5014, Córdoba, Argentina
| |
Collapse
|
13
|
Yang Y, Wu J, Zhou W, Ji G, Dang Y. Protein posttranslational modifications in metabolic diseases: basic concepts and targeted therapies. MedComm (Beijing) 2024; 5:e752. [PMID: 39355507 PMCID: PMC11442990 DOI: 10.1002/mco2.752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/03/2024] Open
Abstract
Metabolism-related diseases, including diabetes mellitus, obesity, hyperlipidemia, and nonalcoholic fatty liver disease, are becoming increasingly prevalent, thereby posing significant threats to human health and longevity. Proteins, as the primary mediators of biological activities, undergo various posttranslational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, methylation, and SUMOylation, among others, which substantially diversify their functions. These modifications are crucial in the physiological and pathological processes associated with metabolic disorders. Despite advancements in the field, there remains a deficiency in contemporary summaries addressing how these modifications influence processes of metabolic disease. This review aims to systematically elucidate the mechanisms through which PTM of proteins impact the progression of metabolic diseases, including diabetes, obesity, hyperlipidemia, and nonalcoholic fatty liver disease. Additionally, the limitations of the current body of research are critically assessed. Leveraging PTMs of proteins provides novel insights and therapeutic targets for the prevention and treatment of metabolic disorders. Numerous drugs designed to target these modifications are currently in preclinical or clinical trials. This review also provides a comprehensive summary. By elucidating the intricate interplay between PTMs and metabolic pathways, this study advances understanding of the molecular mechanisms underlying metabolic dysfunction, thereby facilitating the development of more precise and effective disease management strategies.
Collapse
Affiliation(s)
- Yunuo Yang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Jiaxuan Wu
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Wenjun Zhou
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Guang Ji
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| | - Yanqi Dang
- Institute of Digestive DiseasesChina‐Canada Center of Research for Digestive Diseases (ccCRDD)Shanghai University of Traditional Chinese MedicineShanghaiChina
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine (Shanghai University of Traditional Chinese Medicine)ShanghaiChina
| |
Collapse
|
14
|
Fu Y, Hua Y, Alam N, Liu E. Progress in the Study of Animal Models of Metabolic Dysfunction-Associated Steatotic Liver Disease. Nutrients 2024; 16:3120. [PMID: 39339720 PMCID: PMC11435380 DOI: 10.3390/nu16183120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has recently been proposed as an alternative term to NAFLD. MASLD is a globally recognized chronic liver disease that poses significant health concerns and is frequently associated with obesity, insulin resistance, and hyperlipidemia. To better understand its pathogenesis and to develop effective treatments, it is essential to establish suitable animal models. Therefore, attempts have been made to establish modelling approaches that are highly similar to human diet, physiology, and pathology to better replicate disease progression. Here, we reviewed the pathogenesis of MASLD disease and summarised the used animal models of MASLD in the last 7 years through the PubMed database. In addition, we have summarised the commonly used animal models of MASLD and describe the advantages and disadvantages of various models of MASLD induction, including genetic models, diet, and chemically induced models, to provide directions for research on the pathogenesis and treatment of MASLD.
Collapse
Affiliation(s)
- Yu Fu
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (Y.H.)
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| | - Yuxin Hua
- MOE Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou 730000, China; (Y.F.); (Y.H.)
| | - Naqash Alam
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| | - Enqi Liu
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, 76 Yanta West Road, Xi’an 710061, China;
| |
Collapse
|
15
|
Markou M, Katsouda A, Papaioannou V, Argyropoulou A, Vanioti M, Tamvakopoulos C, Skaltsounis LA, Halabalaki M, Mitakou S, Papapetropoulos A. Anti-obesity effects of Beta vulgaris and Eruca sativa-based extracts. Phytother Res 2024; 38:4757-4773. [PMID: 39120436 DOI: 10.1002/ptr.8291] [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: 02/01/2024] [Revised: 06/29/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024]
Abstract
Obesity is a major source of morbidity worldwide with more than 2 billion adults being overweight or obese. The incidence of obesity has tripled in the last 50 years, leading to an increased risk for a variety of noncommunicable diseases. Previous studies have demonstrated the positive effects of green leafy vegetables on weight gain and obesity and have attributed these beneficial properties, at least in part, to nitrates and isothiocyanates. Nitrates are converted to nitric oxide (NO) and isothiocyanates are known to release hydrogen sulfide (H2S). Herein, we investigated the effect of extracts and fractions produced from Beta vulgaris and Eruca sativa for their ability to limit lipid accumulation, regulate glucose homeostasis, and reduce body weight. Extracts from the different vegetables were screened for their ability to limit lipid accumulation in adipocytes and hepatocytes and for their ability to promote glucose uptake in skeletal muscle cultures; the most effective extracts were next tested in vivo. Wild type mice were placed on high-fat diet for 8 weeks to promote weight gain; animals receiving the selected B. vulgaris and E. sativa extracts exhibited attenuated body weight. Treatment with extracts also led to reduced white adipose tissue depot mass, attenuated adipocyte size, reduced expression of Dgat2 and PPARγ expression, and improved liver steatosis. In contrast, the extracts failed to improve glucose tolerance in obese animals and did not affect blood pressure. Taken together, our data indicate that extracts produced from B. vulgaris and E. sativa exhibit anti-obesity effects, suggesting that dietary supplements containing nitrates and sulfide-releasing compounds might be useful in limiting weight gain.
Collapse
Affiliation(s)
- Maria Markou
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonia Katsouda
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Varvara Papaioannou
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Argyropoulou
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
- PharmaGnose S.A., Oinofyta, Greece
| | - Marianna Vanioti
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantin Tamvakopoulos
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Leandros A Skaltsounis
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Halabalaki
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Sofia Mitakou
- Division of Pharmacognosy and Natural Products Chemistry, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Papapetropoulos
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Laboratory of Pharmacology, Department of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
16
|
Xie L, Hao X, Xie J, Mo J, Yuan C, Chen W. Acetylated pelargonidin-3- O-glucoside alleviates hepatocyte lipid deposition through activating the AMPK-mediated lysosome-autophagy pathway and redox state. Food Funct 2024; 15:6929-6942. [PMID: 38659316 DOI: 10.1039/d4fo00185k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a worldwide public health issue, but a widely accepted therapy is still lacking until now. Anthocyanins are natural flavonoid compounds that possess various bioactivities, but their applications are limited due to their low bioavailability and stability. Acylated anthocyanins are reported to show higher stability, whereas their effects on NAFLD are still unclear. Herein, pelargonidin-3-O-(6''-acetyl)-glucoside (Ace Pg3G) was found to dose-dependently reduce intracellular lipid droplets and triglycerides, and improve cellular oxidative stress that accompanied lipid deposition. Besides, Ace Pg3G was proved to activate AMPK phosphorylation, thus stimulating AMPK-mediated lysosome-autophagy pathway to eliminate overloaded lipid. Further study unveiled that Ace Pg3G regulated genes related to lipid metabolism downstream of AMPK to inhibit lipid synthesis and accelerate lipid oxidation. Overall, this study provided the first evidence, to our best knowledge, that Ace Pg3G ameliorated free fatty acid-induced lipid deposition in hepatocytes through regulating AMPK-mediated autophagy pathways and redox state.
Collapse
Affiliation(s)
- Lianghua Xie
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Xin Hao
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jiahong Xie
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jianling Mo
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
| | - Changzheng Yuan
- School of Public Health, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Wei Chen
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China.
- Department of Food Science and Nutrition, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
17
|
Lin K, Zhang Y, Shen Y, Xu Y, Huang M, Liu X. Hydrogen Sulfide can Scavenge Free Radicals to Improve Spinal Cord Injury by Inhibiting the p38MAPK/mTOR/NF-κB Signaling Pathway. Neuromolecular Med 2024; 26:26. [PMID: 38907170 DOI: 10.1007/s12017-024-08794-1] [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: 04/21/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Spinal cord injury (SCI) causes irreversible cell loss and neurological dysfunctions. Presently, there is no an effective clinical treatment for SCI. It can be the only intervention measure by relieving the symptoms of patients such as pain and fever. Free radical-induced damage is one of the validated mechanisms in the complex secondary injury following primary SCI. Hydrogen sulfide (H2S) as an antioxidant can effectively scavenge free radicals, protect neurons, and improve SCI by inhibiting the p38MAPK/mTOR/NF-κB signaling pathway. In this report, we analyze the pathological mechanism of SCI, the role of free radical-mediated the p38MAPK/mTOR/NF-κB signaling pathway in SCI, and the role of H2S in scavenging free radicals and improving SCI.
Collapse
Affiliation(s)
- Kexin Lin
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yanyang Shen
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yiqin Xu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China.
| |
Collapse
|
18
|
Xu J, Jia W, Zhang G, Liu L, Wang L, Wu D, Tao J, Yue H, Zhang D, Zhao X. Extract of Silphium perfoliatum L. improve lipid accumulation in NAFLD mice by regulating AMPK/FXR signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 327:118054. [PMID: 38484950 DOI: 10.1016/j.jep.2024.118054] [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: 02/12/2024] [Revised: 03/09/2024] [Accepted: 03/12/2024] [Indexed: 03/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Globally, the incidence rate and number of patients with nonalcoholic fatty liver disease are increasing, which has become one of the greatest threats to human health. However, there is still no effective therapy and medicine so far. Silphium perfoliatum L. is a perennial herb native to North America, which is used to improve physical fitness and treat liver and spleen related diseases in the traditional medicinal herbs of Indian tribes. This herb is rich in chlorogenic acids, which have the functions of reducing blood lipids, losing weight and protecting liver. However, the effect of these compounds on nonalcoholic fatty liver disease remains unclear. AIM OF THE STUDY Clarify the therapeutic effects and mechanism of the extract (CY-10) rich in chlorogenic acid and its analogues from Silphium perfoliatum L. on non-alcoholic fatty liver disease, and to determine the active compounds. MATERIALS AND METHODS A free fatty acid-induced steatosis model of HepG2 cells was established to evaluate the in vitro activity of CY-10 in promoting lipid metabolism. Further, a high-fat diet-induced NAFLD model in C57BL/6 mice was established to detect the effects of CY-10 on various physiological and biochemical indexes in mice, and to elucidate the in vivo effects of the extract on regulating lipid metabolism, anti-inflammation and hepatoprotection, and nontarget lipid metabolomics was performed to analyze differential metabolites of fatty acids in the liver. Subsequently, western blotting and immunohistochemistry were used to analyze the target of the extract and elucidate its mechanism of action. Finally, the active compounds in CY-10 were elucidated through in vitro activity screening. RESULTS The results indicated that CY-10 significantly attenuated lipid droplet deposition in HepG2 cells. The results of in vivo experiments showed that CY-10 significantly reduce HFD-induced mouse body weight and organ index, improve biochemical indexes, oxidation levels and inflammatory responses in the liver and serum, thereby protecting the liver tissue. It can promote the metabolism of unsaturated fatty acids in the liver and reduce the generation of saturated fatty acids. Furthermore, it is clarified that CY-10 can promote lipid metabolism balance by regulating AMPK/FXR/SREPB-1c/PPAR-γ signal pathway. Ultimately, the main active compound was proved to be cryptochlorogenic acid, which has a strong promoting effect on the metabolism of fatty acids in cells. Impressively, the activities of CY-10 and cryptochlorogenic acid were stronger than simvastatin in vitro and in vivo. CONCLUSION For the first time, it is clarified that the extract rich in chlorogenic acids and its analogues in Silphium perfoliatum L. have good therapeutic effects on non-alcoholic fatty liver disease. It is confirmed that cryptochlorogenic acid is the main active compound and has good potential for medicine.
Collapse
Affiliation(s)
- Jiyu Xu
- College of Ecological Environmental Engineering, Qinghai University, China.
| | - Wenjing Jia
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, CAS and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Qinghai, 810008, China; University of Chinese Academy of Sciences, China.
| | - Guoying Zhang
- College of Ecological Environmental Engineering, Qinghai University, China.
| | - Liying Liu
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, CAS and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Qinghai, 810008, China; University of Chinese Academy of Sciences, China.
| | - Luya Wang
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, CAS and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Qinghai, 810008, China; University of Chinese Academy of Sciences, China.
| | - Di Wu
- College of Ecological Environmental Engineering, Qinghai University, China.
| | - Jihong Tao
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, CAS and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Qinghai, 810008, China.
| | - Huilan Yue
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, CAS and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Qinghai, 810008, China.
| | - Dejun Zhang
- College of Ecological Environmental Engineering, Qinghai University, China.
| | - Xiaohui Zhao
- Qinghai Key Laboratory of Qinghai-Tibetan Plateau Biological Resources, Northwest Institute of Plateau Biology, CAS and Qinghai Provincial Key Laboratory of Tibetan Medicine Research, Qinghai, 810008, China.
| |
Collapse
|
19
|
Zou Z, Liu X, Yu J, Ban T, Zhang Z, Wang P, Huang R, Zheng F, Chang Y, Peng W, Tang Y, Feng X, Zhao Z, Lv X, Huang S, Guo J, Tuo Y, Zhou Z, Liang S. Nuclear miR-204-3p mitigates metabolic dysfunction-associated steatotic liver disease in mice. J Hepatol 2024; 80:834-845. [PMID: 38331323 DOI: 10.1016/j.jhep.2024.01.029] [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: 04/06/2023] [Revised: 01/16/2024] [Accepted: 01/29/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND & AIMS Accumulating evidence has indicated the presence of mature microRNAs (miR) in the nucleus, but their effects on steatohepatitis remain elusive. We have previously demonstrated that the intranuclear miR-204-3p in macrophages protects against atherosclerosis, which shares multiple risk factors with metabolic dysfunction-associated steatotic liver disease (MASLD). Herein, we aimed to explore the functional significance of miR-204-3p in steatohepatitis. METHODS miR-204-3p levels and subcellular localization were assessed in the livers and peripheral blood mononuclear cells of patients with MASLD. Wild-type mice fed high-fat or methionine- and choline-deficient diets were injected with an adeno-associated virus system containing miR-204-3p to determine the effect of miR-204-3p on steatohepatitis. Co-culture systems were applied to investigate the crosstalk between macrophages and hepatocytes or hepatic stellate cells (HSCs). Multiple high-throughput epigenomic sequencings were performed to explore miR-204-3p targets. RESULTS miR-204-3p expression decreased in livers and macrophages in mice and patients with fatty liver. In patients with MASLD, miR-204-3p levels in peripheral blood mononuclear cells were inversely related to the severity of hepatic inflammation and damage. Macrophage-specific miR-204-3p overexpression reduced steatohepatitis in high-fat or methionine- and choline-deficient diet-fed mice. miR-204-3p-overexpressing macrophages inhibited TLR4/JNK signaling and pro-inflammatory cytokine release, thereby limiting fat deposition and inflammation in hepatocytes and fibrogenic activation in HSCs. Epigenomic profiling identified miR-204-3p as a specific regulator of ULK1 expression. ULK1 transcription and VPS34 complex activation by intranuclear miR-204-3p improved autophagic flux, promoting the anti-inflammatory effects of miR-204-3p in macrophages. CONCLUSIONS miR-204-3p inhibits macrophage inflammation, coordinating macrophage actions on hepatocytes and HSCs to ameliorate steatohepatitis. Macrophage miR-204-3p may be a therapeutic target for MASLD. IMPACT AND IMPLICATIONS Metabolic dysfunction-associated steatotic liver disease (MASLD) is a chronic inflammatory disease ranging from simple steatosis to steatohepatitis. However, the molecular mechanisms underlying the progression of MASLD remain incompletely understood. Here, we demonstrate that miR-204-3p levels in circulating peripheral blood mononuclear cells are negatively correlated with disease severity in patients with MASLD. Nuclear miR-204-3p activates ULK1 transcription and improves autophagic flux, limiting macrophage activation and hepatic steatosis. Our study provides a novel understanding of the mechanism of macrophage autophagy and inflammation in steatohepatitis and suggests that miR-204-3p may act as a potential therapeutic target for MASLD.
Collapse
Affiliation(s)
- Zhaowei Zou
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Xiu Liu
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jie Yu
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China; Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tao Ban
- Department of General Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150081, China; Department of Pharmacology (State Key Laboratory of Frigid Zone Cardiovascular Diseases, Ministry of Science and Technology; The Key Laboratory of Cardiovascular Research, Ministry of Education) at College of Pharmacy, Harbin Medical University, Harbin 150081, China; Heilongjiang Academy of Medical Sciences, Harbin 150081, China
| | - Ziyi Zhang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Peiqi Wang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Renli Huang
- Department of General Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Fuxin Zheng
- Department of General Surgery, Beihai Hospital, Guangxi University of Chinese Medicine, Beihai 536000, China
| | - Yafei Chang
- Faculty of Forensic Medicine, Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Wanli Peng
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yubo Tang
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoqing Feng
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Ziying Zhao
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaofei Lv
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Shuai Huang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jiawei Guo
- Department of Pharmacology, School of Medicine, Yangtze University, Jingzhou 434023, China
| | - Yonghua Tuo
- Department of Neurosurgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Sijia Liang
- Department of Pharmacology, Cardiac and Cerebral Vascular Research Center, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China; Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China.
| |
Collapse
|
20
|
Castellano-Castillo D, Ramos-Molina B, Frutos MD, Arranz-Salas I, Reyes-Engel A, Queipo-Ortuño MI, Cardona F. RNA expression changes driven by altered epigenetics status related to NASH etiology. Biomed Pharmacother 2024; 174:116508. [PMID: 38579398 DOI: 10.1016/j.biopha.2024.116508] [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: 01/11/2024] [Revised: 03/22/2024] [Accepted: 03/27/2024] [Indexed: 04/07/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a growing health problem due to the increased obesity rates, among other factors. In its more severe stage (NASH), inflammation, hepatocellular ballooning and fibrosis are present in the liver, which can further evolve to total liver dysfunction or even hepatocarcinoma. As a metabolic disease, is associated to environmental factors such as diet and lifestyle conditions, which in turn can influence the epigenetic landscape of the cells, affecting to the gene expression profile and chromatin organization. In this study we performed ATAC-sequencing and RNA-sequencing to interrogate the chromatin status of liver biopsies in subjects with and without NASH and its effects on RNA transcription and NASH etiology. NASH subjects showed transcriptional downregulation for lipid and glucose metabolic pathways (e.g., ABC transporters, AMPK, FoxO or insulin pathways). A total of 229 genes were differentially enriched (ATAC and mRNA) in NASH, which were mainly related to lipid transport activity, nuclear receptor-binding, dicarboxylic acid transporter, and PPARA lipid regulation. Interpolation of ATAC data with known liver enhancer regions showed differential openness at 8 enhancers, some linked to genes involved in lipid metabolism, (i.e., FASN) and glucose homeostasis (i.e., GCGR). In conclusion, the chromatin landscape is altered in NASH patients compared to patients without this liver condition. This alteration might cause mRNA changes explaining, at least partially, the etiology and pathophysiology of the disease.
Collapse
Affiliation(s)
- Daniel Castellano-Castillo
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, Málaga 29010, Spain
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia 30120, Spain.
| | - María Dolores Frutos
- General and Digestive System Surgery Department, Virgen de la Arrixaca University Hospital, Murcia 31020, Spain
| | - Isabel Arranz-Salas
- Instituto de Investigación Biomédica de Málaga-Plataforma BIONAND (IBIMA), Virgen de la Victoria University Hospital, Malaga University, 2ª Planta, Campus Teatinos S/N, Málaga 29010, Spain; Department of Human Physiology, Human Histology, Anatomical Pathology and Physical Education, Malaga University, Málaga 29010, Spain; 11 Department of Anatomical Pathology, Virgen de la Victoria Hospital, Málaga, Spain
| | - Armando Reyes-Engel
- Departamento de especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, 29010, Spain
| | - María Isabel Queipo-Ortuño
- Unidad de Gestión Clínica Intercentros de Oncología Médica, Hospitales Universitarios Regional y Virgen de la Victoria, Instituto de Investigación Biomédica de Málaga (IBIMA)-CIMES-UMA, Málaga 29010, Spain; Departamento de especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, 29010, Spain.
| | - Fernando Cardona
- Departamento de especialidades Quirúrgicas, Bioquímica e Inmunología, Facultad de Medicina, Universidad de Málaga, 29010, Spain
| |
Collapse
|
21
|
Wang D, Zhao H, Xing C, Lv B, Wang X, He B. Androgens exacerbate hepatic triglyceride accumulation in rats with polycystic ovary syndrome by downregulating MTTP expression. Endocrine 2024; 84:735-744. [PMID: 37950821 DOI: 10.1007/s12020-023-03590-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/28/2023] [Indexed: 11/13/2023]
Abstract
PURPOSE Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder, which is closely associated with insulin resistance, glucose and lipid metabolism disorders. Patients with PCOS have a significantly higher risk of non-alcoholic fatty liver disease and are associated with hyperandrogenemia (HA). However, the exact mechanism by which HA exacerbates hepatic steatosis in PCOS has not yet been fully elucidated. This work aims to investigate the effects and underlying mechanisms of androgens on hepatic triglyceride (TG) metabolism in rats with PCOS. METHODS Twenty-four female Sprague-Dawley rats were randomly divided into four groups (6 rats/group): control, high-fat diet (HFD), PCOS, and PCOS + flutamide (Flu). Changes in the estrous cycle, liver and ovarian tissue sections, serum total testosterone, serum and liver biochemical indicators, and key enzymes involved in TG metabolism were studied. RESULTS Hepatocyte steatosis and TG accumulation were more evident in the PCOS group than in the control and HFD groups. The PCOS group showed apparent increases in the levels of serum alanine aminotransferase, aspartate aminotransferase, TG, free fatty acid, fasting insulin, and homeostasis model assessment of insulin resistance. Hepatic VLDL and apoB-100 levels decreased in the PCOS group. After Flu was administered to block the actions of androgens, the above abnormalities had been improved. The expression of MTTP was greatly decreased in the PCOS group and significantly increased after Flu administration. CONCLUSION Hepatic steatosis in PCOS rats was correlated with HA. Androgens may exacerbate hepatic TG accumulation by downregulating MTTP expression in PCOS.
Collapse
Affiliation(s)
- Dongxu Wang
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Han Zhao
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Chuan Xing
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China
| | - Bo Lv
- Department of Endocrinology, Dalian Third People's Hospital, Dalian, 116033, PR China
| | - Xiaochen Wang
- Department of Endocrinology, The People's Hospital of Liaoning Province, Shenyang, 110067, PR China
| | - Bing He
- Department of Endocrinology, Shengjing Hospital of China Medical University, Shenyang, 110004, PR China.
| |
Collapse
|
22
|
Miao K, Zhao Y, Xue N. Gkongensin A, an HSP90β inhibitor, improves hyperlipidemia, hepatic steatosis, and insulin resistance. Heliyon 2024; 10:e29367. [PMID: 38655315 PMCID: PMC11036013 DOI: 10.1016/j.heliyon.2024.e29367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 03/29/2024] [Accepted: 04/07/2024] [Indexed: 04/26/2024] Open
Abstract
The prevalence of obesity and its primary associated comorbidities, such as type 2 diabetes and fatty liver disease, has reached epidemic proportions, with no successful treatment available at present. Heat shock protein 90 (HSP90), a crucial chaperone, plays a key role in de novo lipogenesis (DNL) by stabilizing and maintaining sterol regulatory element binding protein (SREBP) activity. Kongensin A (KA), derived from Croton kongensis, inhibits RIP3-mediated necrosis, showing promise as an anti-necrotic and anti-inflammatory agent. It is not yet clear if KA, acting as an HSP90 inhibitor, can enhance hyperlipidemia, hepatic steatosis, and insulin resistance in obese individuals by controlling lipid metabolism. In this study, we first found that KA can potentially decrease lipid content at the cellular level. C57BL/6J mice were given a high-fat diet (HFD) and received KA and lovastatin through oral administration for 7 weeks. KA improved hyperlipidemia, fatty liver, and insulin resistance, as well as reduced body weight in diet-induced obese (DIO) mice, with no significant alteration in food intake. In vitro, KA suppressed DNL and reduced the amounts of mSREBPs. KA promoted mSREBP degradation via the FBW7-mediated ubiquitin-proteasome pathway. KA decreased the level of p-Akt Ser308, and p-GSK3β Ser9 by inhibiting the interaction between HSP90β and Akt. Overall, KA enhanced hyperlipidemia, hepatic steatosis, and insulin resistance by blocking SREBP activity, thereby impacting the FBW7-controlled ubiquitin-proteasome pathway.
Collapse
Affiliation(s)
- Kun Miao
- Department of Hand Surgery, Fuzhou Second General Hospital, 350007, Fuzhou, Fujian, China
| | - Yawei Zhao
- Department of Pharmacy, Jurong Hospital Affiliated to Jiangsu University, Jurong, 212400, Jiangsu, China
| | - Ning Xue
- Department of Acupuncture, Jurong Hospital Affiliated to Jiangsu University, Jurong, 212400, Jiangsu, China
| |
Collapse
|
23
|
Zhao J, Yang T, Yi J, Hu H, Lai Q, Nie L, Liu M, Chu C, Yang J. AP39 through AMPK-ULK1-FUNDC1 pathway regulates mitophagy, inhibits pyroptosis, and improves doxorubicin-induced myocardial fibrosis. iScience 2024; 27:109321. [PMID: 38558936 PMCID: PMC10981016 DOI: 10.1016/j.isci.2024.109321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 01/10/2024] [Accepted: 02/20/2024] [Indexed: 04/04/2024] Open
Abstract
Doxorubicin induces myocardial injury and fibrosis. Still, no effective interventions are available. AP39 is an H2S donor that explicitly targets mitochondria. This study investigated whether AP39 could improve doxorubicin-induced myocardial fibrosis. Doxorubicin induced significant myocardial fibrosis while suppressing mitophagy-related proteins and elevating pyroptosis-related proteins. Conversely, AP39 reverses these effects, enhancing mitophagy and inhibiting pyroptosis. In vitro experiments revealed that AP39 inhibited H9c2 cardiomyocyte pyroptosis, improved doxorubicin-induced impairment of mitophagy, reduced ROS levels, ameliorated the mitochondrial membrane potential, and upregulated AMPK-ULK1-FUNDC1 expression. In contrast, AMPK inhibitor (dorsomorphin) and ULK1 inhibitor (SBI-0206965) reversed AP39 antagonism of doxorubicin-induced FUNDC1-mediated impairment of mitophagy and secondary cardiomyocyte pyroptosis. These results suggest that mitochondria-targeted H2S can antagonize doxorubicin-induced pyroptosis and impaired mitophagy in cardiomyocytes via AMPK-ULK1-FUNDC1 and ameliorated myocardial fibrosis and remodeling.
Collapse
Affiliation(s)
- Junxiong Zhao
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Ting Yang
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jiali Yi
- Department of Cardiology, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Hongmin Hu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Qi Lai
- School of Pharmaceutical Science of University of South China, Hengyang 421000, China
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Liangui Nie
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Maojun Liu
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Chun Chu
- Department of Pharmacy, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| | - Jun Yang
- Department of Cardiology, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421000, China
| |
Collapse
|
24
|
Lee EH, Lee JH, Kim DY, Lee YS, Jo Y, Dao T, Kim KE, Song DK, Seo JH, Seo YK, Seong JK, Moon C, Han E, Kim MK, Ryu S, Shin M, Roh GS, Jung HR, Osborne TF, Ryu D, Jeon TI, Im SS. Loss of SREBP-1c ameliorates iron-induced liver fibrosis by decreasing lipocalin-2. Exp Mol Med 2024; 56:1001-1012. [PMID: 38622198 PMCID: PMC11058876 DOI: 10.1038/s12276-024-01213-2] [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: 10/19/2023] [Revised: 01/10/2024] [Accepted: 02/01/2024] [Indexed: 04/17/2024] Open
Abstract
Sterol regulatory element-binding protein (SREBP)-1c is involved in cellular lipid homeostasis and cholesterol biosynthesis and is highly increased in nonalcoholic steatohepatitis (NASH). However, the molecular mechanism by which SREBP-1c regulates hepatic stellate cells (HSCs) activation in NASH animal models and patients have not been fully elucidated. In this study, we examined the role of SREBP-1c in NASH and the regulation of LCN2 gene expression. Wild-type and SREBP-1c knockout (1cKO) mice were fed a high-fat/high-sucrose diet, treated with carbon tetrachloride (CCl4), and subjected to lipocalin-2 (LCN2) overexpression. The role of LCN2 in NASH progression was assessed using mouse primary hepatocytes, Kupffer cells, and HSCs. LCN2 expression was examined in samples from normal patients and those with NASH. LCN2 gene expression and secretion increased in CCl4-induced liver fibrosis mice model, and SREBP-1c regulated LCN2 gene transcription. Moreover, treatment with holo-LCN2 stimulated intracellular iron accumulation and fibrosis-related gene expression in mouse primary HSCs, but these effects were not observed in 1cKO HSCs, indicating that SREBP-1c-induced LCN2 expression and secretion could stimulate HSCs activation through iron accumulation. Furthermore, LCN2 expression was strongly correlated with inflammation and fibrosis in patients with NASH. Our findings indicate that SREBP-1c regulates Lcn2 gene expression, contributing to diet-induced NASH. Reduced Lcn2 expression in 1cKO mice protects against NASH development. Therefore, the activation of Lcn2 by SREBP-1c establishes a new connection between iron and lipid metabolism, affecting inflammation and HSCs activation. These findings may lead to new therapeutic strategies for NASH.
Collapse
Affiliation(s)
- Eun-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Jae-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Do-Young Kim
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Young-Seung Lee
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yunju Jo
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Tam Dao
- Department of Molecular Cell Biology, Sungkyunkwan University (SKKU) School of Medicine, Suwon, 16419, Republic of Korea
| | - Kyung Eun Kim
- Department of Anatomy, College of Medicine, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Dae-Kyu Song
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Ji Hae Seo
- Department of Biochemistry, School of Medicine, Keimyung University, Daegu, 42601, Republic of Korea
| | - Young-Kyo Seo
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Je Kyung Seong
- Laboratory of Developmental Biology and Genomics, Research Institute for Veterinary Science, BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea
| | - Changjong Moon
- Department of Veterinary Anatomy and Animal Behavior, College of Veterinary Medicine and BK21 FOUR Program, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Eugene Han
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Mi Kyung Kim
- Department of Internal Medicine, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Seungwan Ryu
- Department of Surgery, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Minsang Shin
- Department of Microbiology, School of Medicine, Kyungpook National University, Daegu, 42601, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy, College of Medicine, Institute of Medical Science, Gyeongsang National University, Jinju, 52727, Republic of Korea
| | - Hye Ra Jung
- Department of Pathology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea
| | - Timothy F Osborne
- Institute for Fundamental Biomedical Research, Department of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, St. Petersburg, FL, 33701, USA
| | - Dongryeol Ryu
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Tae-Il Jeon
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, 42601, Republic of Korea.
| |
Collapse
|
25
|
Shen Q, Liu YJ, Qiu TT, Loon K S, Zhou D. Microplastic-induced NAFLD: Hepatoprotective effects of nanosized selenium. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:115850. [PMID: 38290310 DOI: 10.1016/j.ecoenv.2023.115850] [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: 08/25/2023] [Revised: 11/26/2023] [Accepted: 12/15/2023] [Indexed: 02/01/2024]
Abstract
Polystyrene microplastics (MPs) are persistent environmental pollutants commonly encountered in daily human life. Numerous studies have demonstrated their ability to induce liver damage, including oxidative stress, inflammation, and lipid accumulation. However, limited information exists regarding preventive measures against this issue. In our study, we investigated the potential preventive role of selenium nanoparticles (YC-3-SeNPs) derived from Yak-derived Bacillus cereus, a novel nanobiomaterial known for its antioxidant properties and lipid metabolism regulation. Using transcriptomic and metabolomic analyses, we identified key genes and metabolites associated with oxidative stress and lipid metabolism imbalance induced by MPs. Upregulated genes (Scd1, Fasn, Irs2, and Lpin) and elevated levels of arachidonic and palmitic acid accumulation were observed in MP-exposed mice, but not in those exposed to SeNPs. Further experiments confirmed that SeNPs significantly attenuated liver lipid accumulation and degeneration caused by MPs. Histological results and pathway screening validated our findings, revealing that MPs suppressed the Pparα pathway and Nrf2 pathway, whereas SeNPs activated both pathways. These findings suggest that MPs may contribute to the development of nonalcoholic fatty liver disease (NAFLD), while SeNPs hold promise as a future nanobio-product for its prevention.
Collapse
Affiliation(s)
- Qi Shen
- Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei Province, PR China
| | - Yun Jie Liu
- Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei Province, PR China
| | - Tian Tian Qiu
- Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei Province, PR China
| | - San Loon K
- Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei Province, PR China
| | - DongHai Zhou
- Huazhong Agricultural University, Shizishan Street, Hongshan District, Wuhan, Hubei Province, PR China.
| |
Collapse
|
26
|
Shen Z, Hou Y, Zhao G, Tan L, Chen J, Dong Z, Ni C, Pei L. Physiological functions of glucose transporter-2: From cell physiology to links with diabetes mellitus. Heliyon 2024; 10:e25459. [PMID: 38333863 PMCID: PMC10850595 DOI: 10.1016/j.heliyon.2024.e25459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024] Open
Abstract
Glucose is a sugar crucial for human health since it participates in many biochemical reactions. It produces adenosine 5'-triphosphate (ATP) and nucleosides through glucose metabolic and pentose phosphate pathways. These processes require many transporter proteins to assist in transferring glucose across cells, and the most notable ones are glucose transporter-2 (GLUT-2) and sodium/glucose cotransporter 1 (SGLT1). Glucose enters small intestinal epithelial cells from the intestinal lumen by crossing the brush boundary membrane via the SGLT1 cotransporter. It exits the cells by traversing the basolateral membrane through the activity of the GLUT-2 transporter, supplying energy throughout the body. Dysregulation of these glucose transporters is involved in the pathogenesis of several metabolic diseases, such as diabetes. Natural loss of GLUT-2 or its downregulation causes abnormal blood glucose concentrations in the body, such as fasting hypoglycemia and glucose tolerance. Therefore, understanding GLUT-2 physiology is necessary for exploring the mechanisms of diabetes and targeted treatment development. This article reviews how the apical GLUT-2 transporter maintains normal physiological functions of the human body and the adaptive changes this transporter produces under pathological conditions such as diabetes.
Collapse
Affiliation(s)
- Zhean Shen
- Xinjiang Institute of Technology, Aksu, China
| | - Yingze Hou
- Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Guo Zhao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Libi Tan
- School of Laboratory Medicine and Biotechnology, Southern Medical University, China
| | - Jili Chen
- Department of Nutrition and Food Hygiene School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Ziqi Dong
- School of Public Health, Peking University Health Science Center, Beijing 100021, China
| | - Chunxiao Ni
- Hangzhou Lin ‘an District Center for Disease Control and Prevention, Hangzhou, China
| | | |
Collapse
|
27
|
Ren Q, Sun Q, Fu J. Dysfunction of autophagy in high-fat diet-induced non-alcoholic fatty liver disease. Autophagy 2024; 20:221-241. [PMID: 37700498 PMCID: PMC10813589 DOI: 10.1080/15548627.2023.2254191] [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: 05/01/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023] Open
Abstract
ABBREVIATIONS ACOX1: acyl-CoA oxidase 1; ADH5: alcohol dehydrogenase 5 (class III), chi polypeptide; ADIPOQ: adiponectin, C1Q and collagen domain containing; ATG: autophagy related; BECN1: beclin 1; CRTC2: CREB regulated transcription coactivator 2; ER: endoplasmic reticulum; F2RL1: F2R like trypsin receptor 1; FA: fatty acid; FOXO1: forkhead box O1; GLP1R: glucagon like peptide 1 receptor; GRK2: G protein-coupled receptor kinase 2; GTPase: guanosine triphosphatase; HFD: high-fat diet; HSCs: hepatic stellate cells; HTRA2: HtrA serine peptidase 2; IRGM: immunity related GTPase M; KD: knockdown; KDM6B: lysine demethylase 6B; KO: knockout; LAMP2: lysosomal associated membrane protein 2; LAP: LC3-associated phagocytosis; LDs: lipid droplets; Li KO: liver-specific knockout; LSECs: liver sinusoidal endothelial cells; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAP3K5: mitogen-activated protein kinase kinase kinase 5; MED1: mediator complex subunit 1; MTOR: mechanistic target of rapamycin kinase; MTORC1: mechanistic target of rapamycin complex 1; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; NFE2L2: NFE2 like bZIP transcription factor 2; NOS3: nitric oxide synthase 3; NR1H3: nuclear receptor subfamily 1 group H member 3; OA: oleic acid; OE: overexpression; OSBPL8: oxysterol binding protein like 8; PA: palmitic acid; RUBCNL: rubicon like autophagy enhancer; PLIN2: perilipin 2; PLIN3: perilipin 3; PPARA: peroxisome proliferator activated receptor alpha; PRKAA2/AMPK: protein kinase AMP-activated catalytic subunit alpha 2; RAB: member RAS oncogene family; RPTOR: regulatory associated protein of MTOR complex 1; SCD: stearoyl-CoA desaturase; SIRT1: sirtuin 1; SIRT3: sirtuin 3; SNARE: soluble N-ethylmaleimide-sensitive factor attachment protein receptor; SQSTM1/p62: sequestosome 1; SREBF1: sterol regulatory element binding transcription factor 1;SREBF2: sterol regulatory element binding transcription factor 2; STING1: stimulator of interferon response cGAMP interactor 1; STX17: syntaxin 17; TAGs: triacylglycerols; TFEB: transcription factor EB; TP53/p53: tumor protein p53; ULK1: unc-51 like autophagy activating kinase 1; VMP1: vacuole membrane protein 1.
Collapse
Affiliation(s)
- Qiannan Ren
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiming Sun
- International Institutes of Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang, China
- Department of Biochemistry, and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junfen Fu
- Department of Endocrinology, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| |
Collapse
|
28
|
Wang X, Chen Y, Meng H, Meng F. SREBPs as the potential target for solving the polypharmacy dilemma. Front Physiol 2024; 14:1272540. [PMID: 38269061 PMCID: PMC10806128 DOI: 10.3389/fphys.2023.1272540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/26/2023] [Indexed: 01/26/2024] Open
Abstract
The phenomenon of polypharmacy is a common occurrence among older people with multiple health conditions due to the rapid increase in population aging and the popularization of clinical guidelines. The prevalence of metabolic syndrome is growing quickly, representing a serious threat to both the public and the worldwide healthcare systems. In addition, it enhances the risk of cardiovascular disease as well as mortality and morbidity. Sterol regulatory element binding proteins (SREBPs) are basic helix-loop-helix leucine zipper transcription factors that transcriptionally modulate genes that regulate lipid biosynthesis and uptake, thereby serving an essential role in biological systems regulation. In this article, we have described the structure of SREBPs and explored their activation and regulation of signals. We also reveal that SREBPs are intricately involved in the modulation of metabolic diseases and thus have tremendous potential as the novel target for single-drug therapy for multiple diseases.
Collapse
Affiliation(s)
| | | | | | - Fanbo Meng
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis (Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, Jilin Provincial Molecular Biology Research Center for Precision Medicine of Major Cardiovascular Disease, Jilin Provincial Cardiovascular Research Institute), Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
29
|
Nguyen TTP, Nguyen PL, Park SH, Jung CH, Jeon TI. Hydrogen Sulfide and Liver Health: Insights into Liver Diseases. Antioxid Redox Signal 2024; 40:122-144. [PMID: 37917113 DOI: 10.1089/ars.2023.0404] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Significance: Hydrogen sulfide (H2S) is a recently recognized gasotransmitter involved in physiological and pathological conditions in mammals. It protects organs from oxidative stress, inflammation, hypertension, and cell death. With abundant expression of H2S-production enzymes, the liver is closely linked to H2S signaling. Recent Advances: Hepatic H2S comes from various sources, including gut microbiota, exogenous sulfur salts, and endogenous production. Recent studies highlight the importance of hepatic H2S in liver diseases such as nonalcoholic fatty liver disease (NAFLD), liver injury, and cancer, particularly at advanced stages. Endogenous H2S production deficiency is associated with severe liver disease, while exogenous H2S donors protect against liver dysfunction. Critical Issues: However, the roles of H2S in NAFLD, liver injury, and liver cancer are still debated, and its effects depend on donor type, dosage, treatment duration, and cell type, suggesting a multifaceted role. This review aimed to critically evaluate H2S production, metabolism, mode of action, and roles in liver function and disease. Future Direction: Understanding H2S's precise roles and mechanisms in liver health will advance potential therapeutic applications in preclinical and clinical research. Targeting H2S-producing enzymes and exogenous H2S sources, alone or in combination with other drugs, could be explored. Quantifying endogenous H2S levels may aid in diagnosing and managing liver diseases. Antioxid. Redox Signal. 40, 122-144.
Collapse
Affiliation(s)
- Thuy T P Nguyen
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju, Republic of Korea
- Division of Radiation and Genome Stability, Department of Radiation Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Phuc L Nguyen
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju, Republic of Korea
| | - So-Hyun Park
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Chang Hwa Jung
- Aging and Metabolism Research Group, Korea Food Research Institute, Wanju-gun, Republic of Korea
| | - Tae-Il Jeon
- Department of Animal Science, College of Agriculture and Life Science, Chonnam National University, Gwangju, Republic of Korea
| |
Collapse
|
30
|
Le HH, Hagen MW, Louey S, Tavori H, Thornburg KL, Giraud GD, Hinds MT, Barnes AP. Development of a novel Guinea Pig model producing transgenerational endothelial transcriptional changes driven by maternal food restriction and a second metabolic insult of high fat diet. Front Physiol 2023; 14:1266444. [PMID: 37942229 PMCID: PMC10628814 DOI: 10.3389/fphys.2023.1266444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/26/2023] [Indexed: 11/10/2023] Open
Abstract
Developmental programming of chronic adverse cardiovascular health outcomes has been studied both using numerous human populations and an array of animal models. However, the mechanisms that produce transgenerational effects have been difficult to study due to a lack of developmentally relevant models. As such, how increased disease risk is carried to the second generation has been poorly studied. We hypothesized that the endothelium which mediates many acute and chronic vascular inflammatory responses is a key player in these effects, and epidemiological studies implicate transgenerational nutritional effects on endothelial health. To study the mutigenerational effects of maternal undernutrition on offspring endothelial health, we developed a model of transgenerational nutritional stress in guinea pigs, a translationally relevant precocial species with a relatively short lifespan. First- and second-generation offspring were subjected to a high fat diet in adolescence to exacerbate negative cardiovascular health. To assess transcriptional changes, we performed bulk RNA-sequencing in carotid artery endothelial cells, with groups stratified as prenatal control or food restricted, and postnatal control or high fat diet. We detected statistically significant gene alterations for each dietary permutation, some of which were unique to treatments and other transcriptional signatures shared by multiple or all conditions. These findings highlight a core group of genes altered by high fat diet that is shared by all cohorts and a divergence of transgenerational effects between the prenatal ad libitum and dietary restriction groups. This study establishes the groundwork for this model to be used to better understand the interplay of prenatal stress and genetic reprogramming.
Collapse
Affiliation(s)
- Hillary H. Le
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| | - Matthew W. Hagen
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
| | - Samantha Louey
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
| | - Hagai Tavori
- Knight Cardiovascular Institute, Portland, OR, United States
| | - Kent L. Thornburg
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
| | - George D. Giraud
- Center for Developmental Health, Portland, OR, United States
- Knight Cardiovascular Institute, Portland, OR, United States
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, OR, United States
- VA Portland Health Care System, Portland, OR, United States
| | - Monica T. Hinds
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR, United States
- Center for Developmental Health, Portland, OR, United States
| | | |
Collapse
|
31
|
Zhao H, Zhang Y, Fu X, Chen C, Khattak S, Wang H. The double-edged sword role of hydrogen sulfide in hepatocellular carcinoma. Front Pharmacol 2023; 14:1280308. [PMID: 37886126 PMCID: PMC10598729 DOI: 10.3389/fphar.2023.1280308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/02/2023] [Indexed: 10/28/2023] Open
Abstract
With an increasing worldwide prevalence, hepatocellular carcinoma (HCC) is the most common primary malignant tumor of the liver in the world. It is also the primary reason for cancer-related death in the world. The pathogenesis of HCC is complex, such as DNA methylation changes, immune regulatory disorders, cell cycle disorders, chromosomal instability, and so on. Although many studies have been conducted on HCC, the molecular mechanisms of HCC are not completely understood. At present, there is no effective treatment for HCC. Hydrogen sulfide (H2S) has long been regarded as a toxic gas with the smell of rotten eggs, but recent studies have shown that it is an important gasotransmitter along with carbon monoxide (CO) and nitric oxide (NO). Increasing evidence indicates that H2S has multiple biological functions, such as anti-inflammation, anti-apoptosis, anti-oxidative stress, and so on. Recently, a lot of evidence has shown that H2S has a "double-edged sword" effect in HCC, but the mechanism is not fully understood. Here, we reviewed the progress on the role and mechanism of H2S in HCC in recent years, hoping to provide a theoretical reference for future related research.
Collapse
Affiliation(s)
- Huijie Zhao
- Institute of Chronic Disease Risks Assessment, Henan University, Kaifeng, China
| | - Yanting Zhang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
- School of Clinical Medicine, Henan University, Kaifeng, Henan, China
| | - Xiaodi Fu
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| | - Chaoren Chen
- School of Nursing and Health, Institute of Nursing and Health, Henan University, Kaifeng, Henan, China
| | - Saadullah Khattak
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
- School of Life Sciences, Henan University, Kaifeng, China
| | - Honggang Wang
- School of Basic Medical Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
32
|
Petri BJ, Cave MC, Klinge CM. Changes in m6A in Steatotic Liver Disease. Genes (Basel) 2023; 14:1653. [PMID: 37628704 PMCID: PMC10454815 DOI: 10.3390/genes14081653] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Fatty liver disease is one of the major causes of morbidity and mortality worldwide. Fatty liver includes non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH), now replaced by a consensus group as metabolic dysfunction-associated steatotic liver disease (MASLD). While excess nutrition and obesity are major contributors to fatty liver, the underlying mechanisms remain largely unknown and therapeutic interventions are limited. Reversible chemical modifications in RNA are newly recognized critical regulators controlling post-transcriptional gene expression. Among these modifications, N6-methyladenosine (m6A) is the most abundant and regulates transcript abundance in fatty liver disease. Modulation of m6A by readers, writers, and erasers (RWE) impacts mRNA processing, translation, nuclear export, localization, and degradation. While many studies focus on m6A RWE expression in human liver pathologies, limitations of technology and bioinformatic methods to detect m6A present challenges in understanding the epitranscriptomic mechanisms driving fatty liver disease progression. In this review, we summarize the RWE of m6A and current methods of detecting m6A in specific genes associated with fatty liver disease.
Collapse
Affiliation(s)
- Belinda J. Petri
- Department of Biochemistry, University of Louisville School of Medicine, Louisville, KY 40292, USA;
| | - Matthew C. Cave
- Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville, Louisville, KY 40292, USA;
- Hepatobiology and Toxicology Center, University of Louisville, Louisville, KY 40292, USA
- Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40292, USA
| | - Carolyn M. Klinge
- Department of Biochemistry, University of Louisville School of Medicine, Louisville, KY 40292, USA;
- Center for Integrative Environmental Health Sciences (CIEHS), University of Louisville, Louisville, KY 40292, USA;
| |
Collapse
|
33
|
Geng F, Zhong Y, Su H, Lefai E, Magaki S, Cloughesy TF, Yong WH, Chakravarti A, Guo D. SREBP-1 upregulates lipophagy to maintain cholesterol homeostasis in brain tumor cells. Cell Rep 2023; 42:112790. [PMID: 37436895 PMCID: PMC10528745 DOI: 10.1016/j.celrep.2023.112790] [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: 06/27/2022] [Revised: 05/22/2023] [Accepted: 06/23/2023] [Indexed: 07/14/2023] Open
Abstract
Cholesterol is a structural component of cell membranes. How rapidly growing tumor cells maintain membrane cholesterol homeostasis is poorly understood. Here, we found that glioblastoma (GBM), the most lethal brain tumor, maintains normal levels of membrane cholesterol but with an abundant presence of cholesteryl esters (CEs) in its lipid droplets (LDs). Mechanistically, SREBP-1 (sterol regulatory element-binding protein 1), a master transcription factor that is activated upon cholesterol depletion, upregulates critical autophagic genes, including ATG9B, ATG4A, and LC3B, as well as lysosome cholesterol transporter NPC2. This upregulation promotes LD lipophagy, resulting in the hydrolysis of CEs and the liberation of cholesterol from the lysosomes, thus maintaining plasma membrane cholesterol homeostasis. When this pathway is blocked, GBM cells become quite sensitive to cholesterol deficiency with poor growth in vitro. Our study unravels an SREBP-1-autophagy-LD-CE hydrolysis pathway that plays an important role in maintaining membrane cholesterol homeostasis while providing a potential therapeutic avenue for GBM.
Collapse
Affiliation(s)
- Feng Geng
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Yaogang Zhong
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Huali Su
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Etienne Lefai
- Human Nutrition Unit, French National Research Institute for Agriculture, Food and Environment, University Clermont Auvergne, 63122 Clermont-Ferrand, France
| | - Shino Magaki
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | - Timothy F Cloughesy
- Department of Neurology (Neuro-Oncology), David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| | - William H Yong
- Department of Pathology and Laboratory Medicine, School of Medicine at University of California, Irvine, CA 92617, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA
| | - Deliang Guo
- Department of Radiation Oncology, Ohio State Comprehensive Cancer Center, Arthur G. James Cancer Hospital, The Ohio State University, Columbus, OH 43210, USA; Richard J. Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA; College of Medicine at The Ohio State University, Columbus, OH 43210, USA; Center of Cancer Metabolism, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA.
| |
Collapse
|
34
|
Mastino P, Rosati D, de Soccio G, Romeo M, Pentangelo D, Venarubea S, Fiore M, Meliante PG, Petrella C, Barbato C, Minni A. Oxidative Stress in Obstructive Sleep Apnea Syndrome: Putative Pathways to Hearing System Impairment. Antioxidants (Basel) 2023; 12:1430. [PMID: 37507968 PMCID: PMC10376727 DOI: 10.3390/antiox12071430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
INTRODUCTION OSAS is a disease that affects 2% of men and 4% of women of middle age. It is a major health public problem because untreated OSAS could lead to cardiovascular, metabolic, and cerebrovascular complications. The more accepted theory relates to oxidative stress due to intermittent hypoxia, which leads, after an intense inflammatory response through multiple pathways, to endothelial damage. The objective of this study is to demonstrate a correlation between OSAS and hearing loss, the effect of the CPAP on hearing function, and if oxidative stress is also involved in the damaging of the hearing system. METHODS A review of the literature has been executed. Eight articles have been found, where seven were about the correlation between OSAS and the hearing system, and only one was about the CPAP effects. It is noted that two of the eight articles explored the theory of oxidative stress due to intermittent hypoxia. RESULTS All studies showed a significant correlation between OSAS and hearing function (p < 0.05). CONCLUSIONS Untreated OSAS affects the hearing system at multiple levels. Oxidative stress due to intermittent hypoxia is the main pathogenetic mechanism of damage. CPAP has no effects (positive or negative) on hearing function. More studies are needed, with the evaluation of extended high frequencies, the execution of vocal audiometry in noisy environments, and the evaluation of potential biomarkers due to oxidative stress.
Collapse
Affiliation(s)
- Pierluigi Mastino
- Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
| | - Davide Rosati
- Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
| | - Giulia de Soccio
- Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
| | - Martina Romeo
- Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
| | - Daniele Pentangelo
- Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
| | - Stefano Venarubea
- Division of Clinical Pathology, Director of analysis Laboratory of De Lellis Hospital, Viale Kennedy, 02100 Rieti, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Department of Sense Organs DOS, Sapienza University of Rome, Viale del Policlinico 155, 00161 Roma, Italy
| | - Piero Giuseppe Meliante
- Department of Sense Organs DOS, Sapienza University of Rome, Viale del Policlinico 155, 00161 Roma, Italy
| | - Carla Petrella
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Department of Sense Organs DOS, Sapienza University of Rome, Viale del Policlinico 155, 00161 Roma, Italy
| | - Christian Barbato
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Department of Sense Organs DOS, Sapienza University of Rome, Viale del Policlinico 155, 00161 Roma, Italy
| | - Antonio Minni
- Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
- Department of Sense Organs DOS, Sapienza University of Rome, Viale del Policlinico 155, 00161 Roma, Italy
- Clinical Pathology Physician, Director of Analysis Laboratory of De Lellis Hospital, Division of Otolaryngology-Head and Neck Surgery, Ospedale San Camillo de Lellis, ASL Rieti-Sapienza University, Viale Kennedy, 02100 Rieti, Italy
| |
Collapse
|
35
|
Yu S, Song JH, Kim HS, Hong S, Park SK, Park SH, Lee J, Chae YC, Park JH, Lee YG. Patulin alleviates hepatic lipid accumulation by regulating lipogenesis and mitochondrial respiration. Life Sci 2023:121816. [PMID: 37271452 DOI: 10.1016/j.lfs.2023.121816] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/15/2023] [Accepted: 05/24/2023] [Indexed: 06/06/2023]
Abstract
AIMS The aim of this study is to evaluate the effects of patulin on hepatic lipid metabolism and mitochondrial oxidative function and elucidate the underlying molecular mechanisms. MAIN METHODS The effects of patulin on hepatic lipid accumulation were evaluated in free fatty acid-treated AML12 or HepG2 cells through oil red O staining, triglyceride assay, real-time polymerase chain reaction, and western blotting. Alteration of mitochondrial oxidative capacity by patulin treatment was determined using Seahorse analysis to measure the oxygen consumption rate. KEY FINDINGS The increased amounts of lipid droplets induced by free fatty acids were significantly reduced by patulin treatment. Patulin markedly activated the CaMKII/AMP-activated protein kinase (AMPK)/proliferator-activated receptor-γ coactivator (PGC)-1α signaling pathway in hepatocytes, reduced the expression of sterol regulatory element binding protein 1c (SREBP-1c) and lipogenic genes, and increased the expression of genes related to mitochondrial fatty acid oxidation. In addition, patulin treatment enhanced the mitochondrial consumption rate and increased the expression of mitochondrial oxidative phosphorylation proteins in HepG2 hepatocytes. The effects of patulin on anti-lipid accumulation; SREBP-1c, PGC-1α, and carnitine palmitoyltransferase 1 expression; and mitochondrial oxidative capacity were significantly prevented by compound C, an AMPK inhibitor. SIGNIFICANCE Patulin is a potent inducer of the AMPK pathway, and AMPK-mediated mitochondrial activation is required for the efficacy of patulin to inhibit hepatic lipid accumulation. This study is the first to report that patulin is a promising bioactive compound that prevents the development and worsening of fatty liver diseases, including non-alcoholic fatty liver disease, by improving mitochondrial quality and lipid metabolism.
Collapse
Affiliation(s)
- Seungmin Yu
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Ji-Hye Song
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Hee Soo Kim
- Aging and Metabolism Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Seulmin Hong
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Seon Kyeong Park
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Soo Hyun Park
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Jangho Lee
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Young Chan Chae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jae Ho Park
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea
| | - Yu Geon Lee
- Personalized Diet Research Group, Korea Food Research Institute (KFRI), Wanju 55365, Republic of Korea.
| |
Collapse
|
36
|
Lv R, Liu X, Zhang Y, Dong N, Wang X, He Y, Yue H, Yin Q. Pathophysiological mechanisms and therapeutic approaches in obstructive sleep apnea syndrome. Signal Transduct Target Ther 2023; 8:218. [PMID: 37230968 DOI: 10.1038/s41392-023-01496-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 64.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/27/2023] Open
Abstract
Obstructive sleep apnea syndrome (OSAS) is a common breathing disorder in sleep in which the airways narrow or collapse during sleep, causing obstructive sleep apnea. The prevalence of OSAS continues to rise worldwide, particularly in middle-aged and elderly individuals. The mechanism of upper airway collapse is incompletely understood but is associated with several factors, including obesity, craniofacial changes, altered muscle function in the upper airway, pharyngeal neuropathy, and fluid shifts to the neck. The main characteristics of OSAS are recurrent pauses in respiration, which lead to intermittent hypoxia (IH) and hypercapnia, accompanied by blood oxygen desaturation and arousal during sleep, which sharply increases the risk of several diseases. This paper first briefly describes the epidemiology, incidence, and pathophysiological mechanisms of OSAS. Next, the alterations in relevant signaling pathways induced by IH are systematically reviewed and discussed. For example, IH can induce gut microbiota (GM) dysbiosis, impair the intestinal barrier, and alter intestinal metabolites. These mechanisms ultimately lead to secondary oxidative stress, systemic inflammation, and sympathetic activation. We then summarize the effects of IH on disease pathogenesis, including cardiocerebrovascular disorders, neurological disorders, metabolic diseases, cancer, reproductive disorders, and COVID-19. Finally, different therapeutic strategies for OSAS caused by different causes are proposed. Multidisciplinary approaches and shared decision-making are necessary for the successful treatment of OSAS in the future, but more randomized controlled trials are needed for further evaluation to define what treatments are best for specific OSAS patients.
Collapse
Affiliation(s)
- Renjun Lv
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xueying Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Yue Zhang
- Department of Geriatrics, the 2nd Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Na Dong
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Xiao Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Yao He
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Hongmei Yue
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Qingqing Yin
- Department of Geriatric Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China.
| |
Collapse
|
37
|
Shi C, Zhang Z, Xu R, Zhang Y, Wang Z. Contribution of HIF-1α/BNIP3-mediated autophagy to lipid accumulation during irinotecan-induced liver injury. Sci Rep 2023; 13:6528. [PMID: 37085612 PMCID: PMC10121580 DOI: 10.1038/s41598-023-33848-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/19/2023] [Indexed: 04/23/2023] Open
Abstract
Irinotecan is a topoisomerase I inhibitor which has been widely used to combat several solid tumors, whereas irinotecan therapy can induce liver injury. Liver injury generally leads to tissue hypoxia, and hypoxia-inducible factor-1α (HIF-1α), a pivotal transcription factor, mediates adaptive pathophysiological responses to lower oxygen condition. Previous studies have reported a relationship between HIF-1α and autophagy, and autophagy impairment is a common characteristic in a variety of diseases. Here, irinotecan (50 mg/kg) was employed on mice, and HepG2 and L-02 cells were cultured with irinotecan (10, 20 and 40 μM). In vivo study, we found that irinotecan treatment increased final liver index, serum aminotransferase level and hepatic lipid accumulation. Impaired autophagic flux and activation of HIF-1α/BNIP3 pathway were also demonstrated in the liver of irinotecan-treated mice. Moreover, irinotecan treatment significantly deteriorated hepatic oxidative stress, evidenced by increased MDA and ROS contents, as well as decreased GSH-Px, SOD and CAT contents. Interestingly, protein levels of NLRP3, cleaved-caspase 1 and IL-1β were enhanced in the liver of mice injected with irinotecan. In vitro study, irinotecan-treated HepG2 and L-02 cells also showed impaired autophagic flux, while HIF-1α inhibition efficaciously removed the accumulated autophagosomes induced by irinotecan. Additionally, irinotecan treatment aggravated lipid accumulation in HepG2 and L-02 cells, and HIF-1α inhibition reversed the effect of irinotecan. Furthermore, HIF-1α inhibition weakened irinotecan-induced NLRP3 inflammasome activation in HepG2 cells. Taken together, our results suggest that irinotecan induces liver injury by orchestrating autophagy via HIF-1α/BNIP3 pathway, and HIF-1α inhibition could alleviate irinotecan-induced lipid accumulation in HepG2 and L-02 cells, which will provide a new clue and direction for the prevention of side effects of clinical chemotherapy drugs.
Collapse
Affiliation(s)
- Congjian Shi
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No.8, Shangsan Road, Fuzhou, 350007, China
| | - Zhenghong Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No.8, Shangsan Road, Fuzhou, 350007, China
| | - Renfeng Xu
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No.8, Shangsan Road, Fuzhou, 350007, China
| | - Yan Zhang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No.8, Shangsan Road, Fuzhou, 350007, China
| | - Zhengchao Wang
- Provincial Key Laboratory for Developmental Biology and Neurosciences, Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, No.8, Shangsan Road, Fuzhou, 350007, China.
| |
Collapse
|
38
|
Wang X, Cong P, Wang X, Wang Z, Liu B, Xue C, Xu J. Docosahexaenoic acid-acylated astaxanthin monoester ameliorates chronic high-fat diet-induced autophagy dysfunction via ULK1 pathway in the hypothalamus of mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:2378-2388. [PMID: 36606564 DOI: 10.1002/jsfa.12429] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 11/21/2022] [Accepted: 01/06/2023] [Indexed: 06/17/2023]
Abstract
BACKGROUND Dietary astaxanthin (AST) exhibits the ability to resist lipid accumulation and stimulate hepatic autophagy. Natural AST predominantly exists in stable esterified forms. More importantly, in our previous study, docosahexaenoic acid-acylated AST monoester (AST-DHA) possessed better stability, bioavailability, and neuroprotective ability than AST in free and diester form. However, the AST-DHA mechanisms of action in regulating the obese phenotype and autophagy of the central nervous system remain unclear. RESULTS High-fat diet (HFD)-fed C57BL/6J mice were orally administered AST-DHA (50 mg/kg body weight/d) for 3 days or 8 weeks. AST-DHA supplementation alleviated HFD-induced abnormal body weight gain, significantly enhanced autophagy with an increased microtubule-associated protein light chain 3 II/I (LC3II/I) ratio, and reduced the accumulation of p62/sequestosome 1 (SQSTM1) in the hypothalamus rather than in the hippocampus. Mechanistically, AST-DHA effectively promoted autophagy and autophagosome formation, and most notably rescued the HFD-impaired autophagosome-lysosome fusion (indicated by the colocalization of LC3 and LAMP1) by regulating mTOR- and AMPK-induced phosphorylation of ULK1. Consequently, AST-DHA enhanced hypothalamic autophagy, leading to pro-opiomelanocortin (POMC) cleavage to produce alpha-melanocyte-stimulating hormone (α-MSH). CONCLUSIONS This study identified AST-DHA as an enhancer of autophagy that plays a beneficial role in restoring hypothalamic autophagy, and as a new potential therapeutic agent against HFD-induced obesity. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xiaoxu Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Peixu Cong
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Xincen Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Zhigao Wang
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Bin Liu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Changhu Xue
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
- Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jie Xu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| |
Collapse
|
39
|
Oh AR, Jeong Y, Yu J, Minh Tam DT, Kang JK, Jung YH, Im SS, Lee SB, Ryu D, Pajvani UB, Kim K. Hepatocyte Kctd17 Inhibition Ameliorates Glucose Intolerance and Hepatic Steatosis Caused by Obesity-induced Chrebp Stabilization. Gastroenterology 2023; 164:439-453. [PMID: 36402191 PMCID: PMC9975067 DOI: 10.1053/j.gastro.2022.11.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 10/13/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022]
Abstract
BACKGROUND & AIMS Obesity predisposes to type 2 diabetes (T2D) and nonalcoholic fatty liver disease (NAFLD), but underlying mechanisms are incompletely understood. Potassium channel tetramerization domain-containing protein 17 (Kctd17) levels are increased in livers from obese mice and humans. In this study, we investigated the mechanism of increased Kctd17 and whether it is causal to obesity-induced metabolic complications. METHODS We transduced Rosa26-LSL-Cas9 knockin mice with AAV8-TBG-Cre (Control), AAV8-U6-Kctd17 sgRNA-TBG-Cre (L-Kctd17), AAV8-U6-Oga sgRNA-TBG-Cre (L-Oga), or AAV8-U6-Kctd17/Oga sgRNA-TBG-Cre (DKO). We fed mice a high-fat diet (HFD) and assessed for hepatic glucose and lipid homeostasis. We generated Kctd17, O-GlcNAcase (Oga), or Kctd17/Oga-knockout hepatoma cells by CRISPR-Cas9, and Kctd17-directed antisense oligonucleotide to test therapeutic potential in vivo. We analyzed transcriptomic data from patients with NAFLD. RESULTS Hepatocyte Kctd17 expression was increased in HFD-fed mice due to increased Srebp1c activity. HFD-fed L-Kctd17 or Kctd17 antisense oligonucleotide-treated mice show improved glucose tolerance and hepatic steatosis, whereas forced Kctd17 expression caused glucose intolerance and hepatic steatosis even in lean mice. Kctd17 induced Oga degradation, resulting in increasing carbohydrate response element-binding protein (Chrebp) protein, so concomitant Oga knockout negated metabolic benefits of hepatocyte Kctd17 deletion. In patients with NAFLD, KCTD17 messenger RNA was positively correlated with expression of Chrebp target and other lipogenic genes. CONCLUSIONS Srebp1c-induced hepatocyte Kctd17 expression in obesity disrupted glucose and lipid metabolism by stabilizing Chrebp, and may represent a novel therapeutic target for obesity-induced T2D and NAFLD.
Collapse
Affiliation(s)
- Ah-Reum Oh
- Department of Biological Sciences, College of Medicine, Inha University, Incheon, Republic of Korea; Program in Biomedical Science and Engineering, Inha University, Incheon, Republic of Korea; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Yelin Jeong
- Department of Biological Sciences, College of Medicine, Inha University, Incheon, Republic of Korea; Program in Biomedical Science and Engineering, Inha University, Incheon, Republic of Korea; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Junjie Yu
- Department of Medicine, Columbia University, New York, New York
| | - Dao Thi Minh Tam
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Jin Ku Kang
- Department of Medicine, Columbia University, New York, New York
| | - Young Hoon Jung
- Department of Biological Sciences, College of Medicine, Inha University, Incheon, Republic of Korea; Program in Biomedical Science and Engineering, Inha University, Incheon, Republic of Korea; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Sang Bae Lee
- Division of Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Dongryeol Ryu
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Utpal B Pajvani
- Department of Medicine, Columbia University, New York, New York.
| | - KyeongJin Kim
- Department of Biological Sciences, College of Medicine, Inha University, Incheon, Republic of Korea; Program in Biomedical Science and Engineering, Inha University, Incheon, Republic of Korea; Research Center for Controlling Intercellular Communication (RCIC), College of Medicine, Inha University, Incheon, Republic of Korea.
| |
Collapse
|
40
|
He B, Zhang Z, Huang Z, Duan X, Wang Y, Cao J, Li L, He K, Nice EC, He W, Gao W, Shen Z. Protein persulfidation: Rewiring the hydrogen sulfide signaling in cell stress response. Biochem Pharmacol 2023; 209:115444. [PMID: 36736962 DOI: 10.1016/j.bcp.2023.115444] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023]
Abstract
The past few decades have witnessed significant progress in the discovery of hydrogen sulfide (H2S) as a ubiquitous gaseous signaling molecule in mammalian physiology, akin to nitric oxide and carbon monoxide. As the third gasotransmitter, H2S is now known to exert a wide range of physiological and cytoprotective functions in the biological systems. However, endogenous H2S concentrations are usually low, and its potential biologic mechanisms responsible have not yet been fully clarified. Recently, a growing body of evidence has demonstrated that protein persulfidation, a posttranslational modification of cysteine residues (RSH) to persulfides (RSSH) elicited by H2S, is a fundamental mechanism of H2S-mediated signaling pathways. Persulfidation, as a biological switch for protein function, plays an important role in the maintenance of cell homeostasis in response to various internal and external stress stimuli and is also implicated in numerous diseases, such as cardiovascular and neurodegenerative diseases and cancer. In this review, the biological significance of protein persulfidation by H2S in cell stress response is reviewed providing a framework for understanding the multifaceted roles of H2S. A mechanism-guided perspective can help open novel avenues for the exploitation of therapeutics based on H2S-induced persulfidation in the context of diseases.
Collapse
Affiliation(s)
- Bo He
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhe Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhao Huang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Xirui Duan
- Department of Oncology, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Yu Wang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Jiangjun Cao
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Kai He
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Weifeng He
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Army Military Medical University, Chongqing 400038, China.
| | - Wei Gao
- Clinical Genetics Laboratory, Affiliated Hospital & Clinical Medical College of Chengdu University, Chengdu 610081, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, Affiliated Lihuili Hospital, Ningbo University, Ningbo 315040, Zhejiang, China.
| |
Collapse
|
41
|
Roglans N, Fauste E, Bentanachs R, Velázquez AM, Pérez-Armas M, Donis C, Panadero MI, Alegret M, Otero P, Bocos C, Laguna JC. Bempedoic Acid Restores Liver H 2S Production in a Female Sprague-Dawley Rat Dietary Model of Non-Alcoholic Fatty Liver. Int J Mol Sci 2022; 24:ijms24010473. [PMID: 36613916 PMCID: PMC9820553 DOI: 10.3390/ijms24010473] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
We previously demonstrated that treatment with BemA (bempedoic acid), an inhibitor of ATP citrate lyase, significantly reduces fatty liver in a model of liver steatosis (HFHFr-female Sprague-Dawley rat fed a high-fat high-fructose diet). Since the hepatic production of the gasotransmitter H2S is impaired in liver disorders, we were interested in determining if the production of H2S was altered in our HFHFr model and whether the administration of BemA reversed these changes. We used stored liver samples from a previous study to determine the total and enzymatic H2S production, as well as the expression of CBS (cystathionine β-synthase), CSE (cystathionine γ-lyase), and 3MST (3-mercaptopiruvate sulfurtransferase), and the expression/activity of FXR (farnesoid X receptor), a transcription factor involved in regulating CSE expression. Our data show that the HFHFr diet reduces the total and enzymatic production of liver H2S, mainly by decreasing the expression of CBS and CSE. Furthermore, BemA treatment restored H2S production, increasing the expression of CBS and CSE, providing evidence for the involvement of FXR transcriptional activity and the mTORC1 (mammalian target of rapamycin1)/S6K1 (ribosomal protein S6 kinase beta-1)/PGC1α (peroxisome proliferator receptor gamma coactivator1α) pathway.
Collapse
Affiliation(s)
- Núria Roglans
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27–31, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Elena Fauste
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Roger Bentanachs
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27–31, 08028 Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Ana M. Velázquez
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27–31, 08028 Barcelona, Spain
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Madelin Pérez-Armas
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Cristina Donis
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - María I. Panadero
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Marta Alegret
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27–31, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
| | - Paola Otero
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Carlos Bocos
- Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Montepríncipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Juan C. Laguna
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, School of Pharmacy and Food Science, University of Barcelona, Av. Joan XXIII 27–31, 08028 Barcelona, Spain
- Spanish Biomedical Research Centre in Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Institute of Biomedicine, University of Barcelona, 08028 Barcelona, Spain
- Correspondence: ; Tel.: +34-93-402-45-30
| |
Collapse
|
42
|
4-PBA Attenuates Fat Accumulation in Cultured Spotted Seabass Fed High-Fat-Diet via Regulating Endoplasmic Reticulum Stress. Metabolites 2022; 12:metabo12121197. [PMID: 36557235 PMCID: PMC9784988 DOI: 10.3390/metabo12121197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/13/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Excessive fat accumulation is a common phenomenon in cultured fish, which can cause metabolic disease such as fatty liver. However, the relative regulatory approach remains to be explored. Based on this, two feeding trials were conducted. Firstly, fish were fed either a normal-fat diet (NFD) or a high-fat diet (HFD) for eight weeks and sampled at the 2nd, 4th, 6th, and 8th week after feeding (Experiment I). In the first four weeks, fish fed an HFD grew faster than those fed an NFD. Conversely, the body weight and weight gain were higher in the NFD group at the 6th and 8th weeks. Under light and transmission electron microscopes, fat accumulation of the liver was accompanied by an obvious endoplasmic reticulum (ER) swell. Accordingly, the expressions of atf-6, ire-1, perk, eif-2α, atf-4, grp78, and chop showed that ER stress was activated at the 6th and 8th weeks. In Experiment II, 50 mg/kg 4-PBA (an ERs inhibitor) was supplemented to an HFD; this was named the 4-PBA group. Then, fish was fed with an NFD, an HFD, and a 4-PBA diet for eight weeks. As the result, the excessive fat deposition caused by an HFD was reversed by 4-PBA. The expression of ER stress-related proteins CHOP and GRP78 was down-regulated by 4-PBA, and the transmission electron microscope images also showed that 4-PBA alleviated ER stress induced by the feeding of an HFD. Furthermore, 4-PBA administration down-regulated SREBP-1C/ACC/FAS, the critical pathways of fat synthesis. In conclusion, the results confirmed that ER stress plays a contributor role in the fat deposition by activating the SREBP-1C/ACC/FAS pathway. 4-PBA as an ER stress inhibitor could reduce fat deposition caused by an HFD via regulating ER stress.
Collapse
|
43
|
Yin G, Liang H, Sun W, Zhang S, Feng Y, Liang P, Chen S, Liu X, Pan W, Zhang F. Shuangyu Tiaozhi decoction alleviates non-alcoholic fatty liver disease by improving lipid deposition, insulin resistance, and inflammation in vitro and in vivo. Front Pharmacol 2022; 13:1016745. [PMID: 36506575 PMCID: PMC9727266 DOI: 10.3389/fphar.2022.1016745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/07/2022] [Indexed: 11/24/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases worldwide. Our previous studies have found that Shuangyu Tiaozhi Decoction (SYTZD) could produce an improvement in NAFLD-related indicators, but the underlying mechanism associated with this improvement remains unclear. The study aimed to investigate the potential mechanism of SYTZD against NAFLD through network pharmacology and experimental verification. The components of SYTZD and SYTZD drug containing serum were analyzed using ultra-performance liquid chromatography to quadrupole/time-of-flight mass spectrometry (UPLC-Q/TOF-MS). Active components and targets of SYTZD were screened by the traditional Chinese medical systems pharmacology (TCMSP) and encyclopedia of traditional Chinese medicine (ETCM) databases. NAFLD-related targets were collected from the GeneCards and DisGeNET databases. The component-disease targets were mapped to identify the common targets of SYTZD against NAFLD. Protein-protein interaction (PPI) network of the common targets was constructed for selecting the core targets. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis of the core targets was performed using the database for annotation, visualization, and integrated discovery (DAVID) database. Furthermore, animal and cell models were constructed for validating the predictions of network pharmacology. Lipid accumulation, liver histopathology, insulin resistance, and core gene expression were measured by oil red O staining, hematoxylin and eosin staining, insulin tolerance test, real-time quantitative polymerase chain reaction, and Western blotting, respectively. Two components and 22 targets of SYTZD against NAFLD were identified by UPLC-Q/TOF-MS and relevant databases. PPI analysis found that ESR1, FASN, mTOR, HIF-1α, VEGFA, and GSK-3β might be the core targets of SYTZD against NAFLD, which were mainly enriched in the thyroid hormone pathway, insulin resistance pathway, HIF-1 pathway, mTOR pathway, and AMPK pathway. Experimental results revealed that SYTZD might exert multiple anti-NAFLD mechanisms, including improvements in lipid deposition, inflammation, and insulin resistance. SYTZD treatment led to decreases in the lipid profiles, hepatic enzyme levels, inflammatory cytokines, and homeostatic model assessment for insulin resistance (HOMA-IR). SYTZD treatment affected relative mRNA and protein levels associated with various pathways. Our findings reveal that SYTZD could alleviate NAFLD through a multi-component, multi-target, and multi-pathway mechanism of action.
Collapse
Affiliation(s)
- Guoliang Yin
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongyi Liang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenxiu Sun
- Department of Nursing, Taishan Vocational College of Nursing, Taian, China
| | - Shizhao Zhang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanan Feng
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Pengpeng Liang
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Suwen Chen
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiangyi Liu
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenchao Pan
- The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Fengxia Zhang,
| |
Collapse
|
44
|
Lee JH, Im SS. Function of gaseous hydrogen sulfide in liver fibrosis. BMB Rep 2022; 55:481-487. [PMID: 36195563 PMCID: PMC9623240 DOI: 10.5483/bmbrep.2022.55.10.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/07/2022] [Accepted: 09/21/2022] [Indexed: 11/06/2022] Open
Abstract
Over the past few years, hydrogen sulfide (H2S) has been shown to exert several biological functions in mammalian. The endogenous production of H2S is mainly mediated by cystathione β-synthase, cystathione γ-lyase and 3-mercaptopyruvate sulfur transferase. These enzymes are broadly expressed in liver tissue and regulates liver function by working on a variety of molecular targets. As an important regulator of liver function, H2S is critically involved in the pathogenesis of various liver diseases, such as non-alcoholic steatohepatitis and liver cancer. Targeting H2S-generating enzymes may be a therapeutic strategy for controlling liver diseases. This review described the function of H2S in liver disease and summarized recent characterized role of H2S in several cellular process of the liver. [BMB Reports 2022; 55(10): 481-487].
Collapse
Affiliation(s)
- Jae-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu 42601, Korea
| |
Collapse
|
45
|
Pu J. Targeting the lysosome: Mechanisms and treatments for nonalcoholic fatty liver disease. J Cell Biochem 2022; 123:1624-1633. [PMID: 35605052 PMCID: PMC9617749 DOI: 10.1002/jcb.30274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/24/2022] [Accepted: 05/02/2022] [Indexed: 11/11/2022]
Abstract
The multiple functions of the lysosome, including degradation, nutrient sensing, signaling, and gene regulation, enable the lysosome to regulate lipid metabolism at different levels. In this review, I summarize the recent studies on lysosomal regulation of lipid metabolism and the alterations of the lysosome functions in the livers affected by nonalcoholic fatty liver disease (NAFLD). NAFLD is a highly prevalent lipid metabolic disorder. The progression of NAFLD leads to nonalcoholic steatohepatitis (NASH) and other severe liver diseases, and thus the prevention and treatments of NAFLD progression are critically needed. Targeting the lysosome is a promising strategy. I also discuss the current manipulations of the lysosome functions in the preclinical studies of NAFLD and propose my perspectives on potential future directions.
Collapse
Affiliation(s)
- Jing Pu
- Department of Molecular Genetics and Microbiology, Autophagy, Inflammation, and Metabolism (AIM) Center of Biomedical Research Excellence, University of New Mexico, Albuquerque, New Mexico, USA
| |
Collapse
|
46
|
Cheng Q, Zhang J, Fang J, Ding H, Xu Y, Lu X, Zhang W. Untargeted metabolomics reveals the role of AQP9 in nonalcoholic fatty liver disease in a mice model. Int J Biol Macromol 2022; 219:864-875. [PMID: 35961555 DOI: 10.1016/j.ijbiomac.2022.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022]
Abstract
Previous studies have shown that AQP9 plays an important role in energy metabolism in nonalcoholic fatty liver disease (NAFLD). Recently, metabolomic analyses were used to determine the slight changes in metabolic profiles and helped to understand the disease progression, therapeutic intervention of NAFLD. A mouse model of NAFLD was established with a high-fat diet (HFD), and Aqp9 knockout mice were constructed. Untargeted metabolomics techniques were used to evaluate the potential mechanism of the effect of AQP9 in NAFLD. The results indicated that AQP9 plays a regulatory role in the occurrence of NAFLD. Moreover, a total of 220 candidate biomarkers were screened and identified. Cluster analysis and enrichment analysis of differential metabolites indicated that fatty acid biosynthesis was mainly disturbed when compared against the control group, which was mitigated by knockout of Aqp9. These results show that untargeted metabolomics help to understand the effects of AQP9 in NAFLD.
Collapse
Affiliation(s)
- Quancheng Cheng
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Junwei Zhang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jinyu Fang
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Huiru Ding
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yiyao Xu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Xin Lu
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Weiguang Zhang
- Department of Human Anatomy and Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
47
|
Zhang Y, Ye S, Lu W, Zhong J, Leng Y, Yang T, Luo J, Xu W, Zhang H, Kong L. RNA helicase DEAD-box protein 5 alleviates nonalcoholic steatohepatitis progression via tethering TSC complex and suppressing mTORC1 signaling. Hepatology 2022; 77:1670-1687. [PMID: 35796622 DOI: 10.1002/hep.32651] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/01/2022] [Accepted: 07/05/2022] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Nonalcoholic fatty liver disease and its progressive form, nonalcoholic steatohepatitis (NASH), are rapidly becoming the top causes of hepatocellular carcinoma (HCC). Currently, there are no approved therapies for the treatment of NASH. DEAD-box protein 5 (DDX5) plays important roles in different cellular processes. However, the precise role of DDX5 in NASH remains unclear. APPROACH AND RESULTS DDX5 expression was downregulated in patients with NASH, mouse models with diet-induced NASH (high-fat diet [HFD], methionine- and choline-deficient diet, and choline-deficient HFD), mouse models with NASH-HCC (diethylnitrosamine with HFD), and palmitic acid-stimulated hepatocytes. Adeno-associated virus-mediated DDX5 overexpression ameliorates hepatic steatosis and inflammation, whereas its deletion worsens such pathology. The untargeted metabolomics analysis was carried out to investigate the mechanism of DDX5 in NASH and NASH-HCC, which suggested the regulatory effect of DDX5 on lipid metabolism. DDX5 inhibits mechanistic target of rapamycin complex 1 (mTORC1) activation by recruiting the tuberous sclerosis complex (TSC)1/2 complex to mTORC1, thus improving lipid metabolism and attenuating the NACHT-, leucine-rich-repeat (LRR)-, and pyrin domain (PYD)-containing protein 3 inflammasome activation. We further identified that the phytochemical compound hyperforcinol K directly interacted with DDX5 and prevented its ubiquitinated degradation mediated by ubiquitin ligase (E3) tripartite motif protein 5, thereby significantly reducing lipid accumulation and inflammation in a NASH mouse model. CONCLUSIONS These findings provide mechanistic insight into the role of DDX5 in mTORC1 regulation and NASH progression, as well as suggest a number of targets and a promising lead compound for therapeutic interventions against NASH.
Collapse
Affiliation(s)
- Yanqiu Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shengtao Ye
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Weijia Lu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jiawen Zhong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yingrong Leng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ting Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jun Luo
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenjun Xu
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hao Zhang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Lingyi Kong
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
48
|
Osborne TF, Espenshade PJ. Lipid balance must be just right to prevent development of severe liver damage. J Clin Invest 2022; 132:e160326. [PMID: 35642642 PMCID: PMC9151688 DOI: 10.1172/jci160326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major health concern that often associates with obesity and diabetes. Fatty liver is usually a benign condition, yet a fraction of individuals progress to severe forms of liver damage, including nonalcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC). Elevated sterol regulatory element-binding protein-driven (SREBP-driven) hepatocyte lipid synthesis is associated with NAFLD in humans and mice. In this issue of the JCI, Kawamura, Matsushita, et al. evaluated the role of SREBP-dependent lipid synthesis in the development of NAFLD, NASH, and HCC in the phosphatase and tensin homolog-knockout (PTEN-knockout) NASH model. Deletion of the gene encoding SREBP cleavage-activating protein (SCAP) from the liver resulted in decreased hepatic lipids, as expected. However, SCAP deletion accelerated progression to more severe liver damage, including NASH and HCC. This study provides a note of caution for those pursuing de novo fat biosynthesis as a therapeutic intervention in human NASH.
Collapse
Affiliation(s)
- Timothy F. Osborne
- Departments of Medicine, Pediatrics, and Biological Chemistry, and Institute for Fundamental Biomedical Research, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - Peter J. Espenshade
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
49
|
Zou L, Liao M, Zhen Y, Zhu S, Chen X, Zhang J, Hao Y, Liu B. Autophagy and beyond: Unraveling the complexity of UNC-51-like kinase 1 (ULK1) from biological functions to therapeutic implications. Acta Pharm Sin B 2022; 12:3743-3782. [PMID: 36213540 PMCID: PMC9532564 DOI: 10.1016/j.apsb.2022.06.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/27/2022] [Accepted: 06/02/2022] [Indexed: 12/13/2022] Open
Abstract
UNC-51-like kinase 1 (ULK1), as a serine/threonine kinase, is an autophagic initiator in mammals and a homologous protein of autophagy related protein (Atg) 1 in yeast and of UNC-51 in Caenorhabditis elegans. ULK1 is well-known for autophagy activation, which is evolutionarily conserved in protein transport and indispensable to maintain cell homeostasis. As the direct target of energy and nutrition-sensing kinase, ULK1 may contribute to the distribution and utilization of cellular resources in response to metabolism and is closely associated with multiple pathophysiological processes. Moreover, ULK1 has been widely reported to play a crucial role in human diseases, including cancer, neurodegenerative diseases, cardiovascular disease, and infections, and subsequently targeted small-molecule inhibitors or activators are also demonstrated. Interestingly, the non-autophagy function of ULK1 has been emerging, indicating that non-autophagy-relevant ULK1 signaling network is also linked with diseases under some specific contexts. Therefore, in this review, we summarized the structure and functions of ULK1 as an autophagic initiator, with a focus on some new approaches, and further elucidated the key roles of ULK1 in autophagy and non-autophagy. Additionally, we also discussed the relationships between ULK1 and human diseases, as well as illustrated a rapid progress for better understanding of the discovery of more candidate small-molecule drugs targeting ULK1, which will provide a clue on novel ULK1-targeted therapeutics in the future.
Collapse
Affiliation(s)
- Ling Zou
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minru Liao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yongqi Zhen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shiou Zhu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiya Chen
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jin Zhang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Corresponding authors. Tel./fax: +86 28 85503817.
| | - Yue Hao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen 518060, China
- Corresponding authors. Tel./fax: +86 28 85503817.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Corresponding authors. Tel./fax: +86 28 85503817.
| |
Collapse
|
50
|
Li T, Wang H, Dong S, Liang M, Ma J, Jiang X, Yu W. Protective effects of maslinic acid on high fat diet-induced liver injury in mice. Life Sci 2022; 301:120634. [PMID: 35568228 DOI: 10.1016/j.lfs.2022.120634] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 11/30/2022]
Abstract
AIMS Due to the prevalence of high-fat diets and lack of exercise, diseases related to nutrient metabolism such as nonalcoholic fatty liver disease (NAFLD) have become one of the reasons causes endangering human liver health. Maslinic acid (MA) is a pentacyclic triterpenoid acid that is abundant in fruits such as hawthorn and jujube. In this study, we investigated the effect of MA on NAFLD to inform the development of dietary supplements for the treatment and prevention of NAFLD. MATERIALS AND METHODS The NAFLD model was established by feeding mice a high-fat diet (HFD). HEPG2 cells were treated with oleic acid and used as a cell culture model. Testing kits, haematoxylin and eosin staining, oil red O staining, western blotting, and immunofluorescence were performed with in vivo and in vitro experiments. KEY FINDINGS The current study revealed that MA significantly reduced liver weight, body weight and serum lipid levels, and protected against liver steatosis and injury induced by a HFD. MA increased the expression of Beclin1, ATG1, and Bcl-2 mRNA and protein while decreasing the expression of TNF-α and IL-1β, caspase-3 and Bax mRNA and protein. Beclin1, and ATG1 were obviously increased, and the mRNA and protein expression of TNF-α and IL-1β were obviously reduced, the mRNA and protein expression of Caspase-3 and Bax were obviously reduced, and the mRNA and protein expression of Bax were obviously increased by MA. SIGNIFICANCE MA reduces the content of fat in the liver cells of NAFLD mice through lipophagy activitiy and reduces inflammation and apoptosis injury.
Collapse
Affiliation(s)
- Tianqi Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Huan Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Siyu Dong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Meng Liang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jun Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - XiaoWen Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| | - Wenhui Yu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|