1
|
Yang H, Xu C, Song J, Li J, Zhang C, Teng C, Ma K, Xie F. Toxicokinetic and liver proteomic study of the Chinese rare minnow (Gobiocypris rarus) exposed to F-53B. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2025; 282:107312. [PMID: 40107147 DOI: 10.1016/j.aquatox.2025.107312] [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: 12/30/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/22/2025]
Abstract
Perfluorooctane sulfonic acid (PFOS) and its alternative 6:2 chlorinated polyfluoroethersulfonate (6:2 Cl-PFESA, also known as F-53B), are frequently detected in a variety of environmental and human samples. These substances have been associated with hepatotoxic effects, including disorders in lipid metabolism and oxidative stress. However, the molecular mechanisms underlying the causal relationship between exposure to F-53B and hepatotoxicity remain inadequately understood. This study investigated the toxicokinetics and mechanisms of hepatotoxicity associated with prolonged exposure to F-53B in adult Chinese rare minnows. Specifically, 5-month-old adult Chinese rare minnow was exposed to concentrations of 10 μg/L and 200 μg/L of F-53B for a duration of 28 days for bioaccumulation assessment, followed by a 14-day period for metabolic evaluation. The findings indicated that the bioaccumulation of F-53B in the tissues was positively correlated with the exposure concentrations. The logarithmic bioconcentration factor (Log BCF28d) was determined to be 2.67 ± 0.02 for the low concentration group and 2.27 ± 0.01 for the high concentration group. The calculated half-lives (t1/2) were 18.50 ± 1.67 days and 21.38 ± 0.31 days for the respective concentration groups. F-53B protein exhibited a distinct tissue-specific distribution in adult Chinese rare minnow, with the following order of enrichment: Blood > Liver > Gonad > Gill > Intestine > Brain > Muscle. F-53B was primarily concentrated in the blood and liver, where the protein content was significantly higher. Exposure to F-53B for 28 days significantly elevated biochemical levels associated with lipid metabolism and increased the activities of the enzymes FAS, PPARα, and ACC in the liver. This exposure also resulted in impairment of the hepatic oxidative system in the Chinese rare minnow, with F-53B significantly reducing most of the measured markers related to oxidative stress (e.g., GSH, SOD, CAT, and MDA). Proteomic analysis indicated that the toxicity of F-53B regulates the expression of proteins across several functional classes. Based on the functional information of the differential proteins provided in UniProt and KEGG, they were categorized into five main categories: Cellular Processes, Environmental Information Processing, Genetic Information Processing, Metabolism, and Organismal Systems. This study indicate that F-53B is bioaccumulative and persistent in Chinese rare minnow, and can further induce oxidative stress and lipid metabolism disorders. Combined with proteomic research methods, the toxicological effects of F-53B on Chinese rare minnow can be better explained.
Collapse
Affiliation(s)
- Hongbo Yang
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Guizhou Academy of Testing and Analysis, Guiyang 550014, China.
| | - Chan Xu
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Guizhou Academy of Testing and Analysis, Guiyang 550014, China
| | - Jieyu Song
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Guizhou Academy of Testing and Analysis, Guiyang 550014, China
| | - Jin Li
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China; Guizhou Academy of Testing and Analysis, Guiyang 550014, China
| | - Chan Zhang
- School of Public Health, the key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 561113, China
| | - Chunli Teng
- Guizhou Jiandee Technology Co., Ltd, Guiyang 550016, China
| | - Kai Ma
- Guizhou Academy of Testing and Analysis, Guiyang 550014, China
| | - Feng Xie
- Guizhou Academy of Testing and Analysis, Guiyang 550014, China; Department of Food science and Engineering Moutai Institute, Renhuai 564507, China
| |
Collapse
|
2
|
Shao C, Lan W, Ding Y, Ye L, Huang J, Liang X, He Y, Zhang J. JTCD attenuates HF by inhibiting activation of HSCs through PPARα-TFEB axis-mediated lipophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156501. [PMID: 39978277 DOI: 10.1016/j.phymed.2025.156501] [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: 10/10/2024] [Revised: 12/28/2024] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Hepatic fibrosis (HF) is an intermediate stage in the progression of chronic liver disease to cirrhosis and has been shown to be a reversible pathological process. Known evidence suggests that activation of hepatic stellate cells (HSCs) and degradation of their lipid droplets (LDs) play an indispensable role in the process of HF. Jiawei Taohe Chengqi Decoction (JTCD) can inhibit the activation of HSCs in the process of HF, but the exact mechanism remains to be elucidated. PURPOSE The aim of this study is to determine whether JTCD inhibits lipophagy and to explore the possible mechanisms of its HF effect in HSCs by regulating the PPARα/TFEB axis. METHODS Network pharmacology and molecular docking were firstly applied to predict the potential mechanism of JTCD for the treatment of HF. In vivo, a mouse model of HF was constructed using carbon tetrachloride (CCl4) solution, and the efficacy of JTCD was assessed by staining of pathological sections, oil red O staining, immunofluorescence (IF), immunohistochemistry (IHC) staining, Western blotting and qRT-PCR. The intervention of JTCD was verified in vitro by induction of activated LX-2 cells with TGF-β solution and intervention using agonists and antagonists of PPARα. Finally, transient transfection of cells using TFEB siRNA was performed for validation studies. RESULTS JTCD effectively alleviated CCl4-induced HF in mice and reduced the levels of HF markers α-smooth muscle actin (α-SMA) and collagen I (COL1A1), and inhibited PPARα expression and lipophagy process. In vitro, JTCD delayed the degradation of LDs and reduced lipophagy in LX-2 cells, suggesting a mechanism involving PPARα/TFEB axis signaling regulation.
Collapse
Affiliation(s)
- Chang Shao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wenfang Lan
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ying Ding
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Linmao Ye
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jiaxin Huang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xiaofan Liang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yi He
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Junjie Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
3
|
Zhang QR, Dong Y, Fan JG. Early-life exposure to gestational diabetes mellitus predisposes offspring to pediatric nonalcoholic fatty liver disease. Hepatobiliary Pancreat Dis Int 2025; 24:128-137. [PMID: 38195352 DOI: 10.1016/j.hbpd.2023.12.007] [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: 08/05/2023] [Accepted: 12/28/2023] [Indexed: 01/11/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the prevailing chronic liver disease in the pediatric population due to the global obesity pandemic. Evidence shows that prenatal and postnatal exposure to maternal abnormalities leads to a higher risk of pediatric NAFLD through persistent alterations in developmental programming. Gestational diabetes mellitus (GDM) is a hyperglycemic syndrome which has become the most prevalent complication in pregnant women. An increasing number of both epidemiologic investigations and animal model studies have validated adverse and long-term outcomes in offspring following GDM exposure in utero. Similarly, GDM is considered a crucial risk factor for pediatric NAFLD. This review aimed to summarize currently published studies concerning the inductive roles of GDM in offspring NAFLD development during childhood and adolescence. Dysregulations in hepatic lipid metabolism and gut microbiota in offspring, as well as dysfunctions in the placenta are potential factors in the pathogenesis of GDM-associated pediatric NAFLD. In addition, potentially effective interventions for GDM-associated offspring NAFLD are also discussed in this review. However, most of these therapeutic approaches still require further clinical research for validation.
Collapse
Affiliation(s)
- Qian-Ren Zhang
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Yan Dong
- Department of Endocrinology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Institute for Pediatric Research, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Jian-Gao Fan
- Center for Fatty Liver, Department of Gastroenterology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; Shanghai Key Lab of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China.
| |
Collapse
|
4
|
Chan P, Cheung PHH, Kang XZ, Cheng Y, Wong CM, Jin DY, Chan CP. Cgref1 is a CREB-H-regulated hepatokine that promotes hepatic de novo lipogenesis by mediating epididymal fat insulin resistance. Int J Biol Sci 2025; 21:2568-2588. [PMID: 40303310 PMCID: PMC12035884 DOI: 10.7150/ijbs.97008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 03/10/2025] [Indexed: 05/02/2025] Open
Abstract
Rationale: Type 2 diabetes mellitus and metabolic dysfunction-associated steatotic liver disease (MASLD) are interrelated metabolic disorders that pose significant health concerns. Hepatokines and other regulatory factors implicated in these diseases are incompletely understood. Here, we report on a new hepatokine named cell growth regulator with EF-hand domain 1 (Cgref1) that modulates lipid metabolism to aggravate these conditions. Methods: Cgref1 was identified by microarray analysis of downregulated genes in liver of Creb3l3 -/- mice. Cgref1 -/- mice were subjected to transcriptomic, metabolomic and lipidomic analyses as well as metabolic assays. Gain-of-function and loss-of-function assays were performed in primary hepatocytes and cultured human and mouse cells. Results: Cgref1 expression is induced by hepatic transcription factor CREB-H. Secreted Cgref1 primarily targets epididymal white adipose tissue (eWAT), where insulin signalling and glucose uptake are suppressed. Cgref1-/- mice showed lower tendencies of developing obesity, hyperglycaemia and dyslipidaemia, associated with compromised hepatic de novo lipogenesis. Thus, Cgref1 poses an advantage to maintain the normal functioning of vital organs by preserving glucose from being absorbed into eWAT. However, in circumstances where Cgref1 expression becomes excessive, eWAT develops insulin resistance, which in turn promotes hepatic glucose production, lipogenesis and MASLD development. Conclusion: As a hepatokine that affects blood glucose levels and lipogenesis, Cgref1 is a potential target in the intervention of metabolic disorders.
Collapse
Affiliation(s)
- Pearl Chan
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Pak-Hin Hinson Cheung
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Xiao-Zhuo Kang
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Yun Cheng
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Chi-Ming Wong
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, 11 Yuk Choi Road, Hung Hom, Hong Kong
| | - Dong-Yan Jin
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| | - Chi-Ping Chan
- School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong
| |
Collapse
|
5
|
Park SY, Cho Y, Son SM, Hur JH, Kim Y, Oh H, Lee HY, Jung S, Park S, Kim IY, Lee SJ, Choi CS. Activin E is a new guardian protecting against hepatic steatosis via inhibiting lipolysis in white adipose tissue. Exp Mol Med 2025; 57:466-477. [PMID: 39948368 PMCID: PMC11873131 DOI: 10.1038/s12276-025-01403-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/21/2024] [Accepted: 12/11/2024] [Indexed: 03/04/2025] Open
Abstract
Hepatic endoplasmic reticulum (ER) stress is implicated in the development of steatosis and its progression to nonalcoholic steatohepatitis (NASH). The ER in the liver can sustain metabolic function by activating defense mechanisms that delay or prevent the progression of nonalcoholic fatty liver disease (NAFLD). However, the precise mechanisms by which the ER stress response protects against NAFLD remain largely unknown. Recently, activin E has been linked to metabolic diseases such as insulin resistance and NAFLD. However, the physiological conditions and regulatory mechanisms driving hepatic Inhbe expression (which encodes activin E) as well as the metabolic role of activin E in NAFLD require further investigation. Here we found that hepatic Inhbe expression increased under prolonged fasting and ER stress conditions, which was mediated by ATF4, as determined by promoter analysis in a mouse model. Consistently, a positive correlation between INHBE and ATF4 expression levels in relation to NAFLD status was confirmed using public human NAFLD datasets. To investigate the role of activin E in hepatic steatosis, we assessed the fluxes of the lipid metabolism in an Inhbe-knockout mouse model. These mice displayed a lean phenotype but developed severe hepatic steatosis under a high-fat diet. The deficiency of Inhbe resulted in increased lipolysis in adipose tissue, leading to increased fatty acid influx into the liver. Conversely, hepatic overexpression of Inhbe ameliorated hepatic steatosis by suppressing lipolysis in adipose tissue through ALK7-Smad signaling. In conclusion, activin E serves as a regulatory hepatokine that prevents fatty acid influx into the liver, thereby protecting against NAFLD.
Collapse
Affiliation(s)
- Shi-Young Park
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Gachon Biomedical Convergence Institute, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Yoonil Cho
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Sae-Mi Son
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
| | - Jang Ho Hur
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, Republic of Korea
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
| | - Hyunhee Oh
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Gachon Biomedical Convergence Institute, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Hui-Young Lee
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Division of Molecular Medicine, Department of Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Sungwon Jung
- Department of Genome Medicine and Science, Gachon University College of Medicine, Incheon, Republic of Korea
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Sanghee Park
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Department of Exercise Rehabilitation, Gachon University, Incheon, Republic of Korea
| | - Il-Young Kim
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Integrative Metabolic Fluxomics Lab, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea
- Division of Molecular Medicine, Department of Medicine, Gachon University College of Medicine, Incheon, Republic of Korea
| | - Se-Jin Lee
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon, Republic of Korea.
- Division of Molecular Medicine, Department of Medicine, Gachon University College of Medicine, Incheon, Republic of Korea.
- Endocrinology, Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea.
| |
Collapse
|
6
|
Tang R, Xie C, Zhang X. NOD1: a metabolic modulator. Front Endocrinol (Lausanne) 2025; 15:1484829. [PMID: 39906040 PMCID: PMC11790428 DOI: 10.3389/fendo.2024.1484829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Nucleotide-binding oligomerization domain 1 (NOD1) is an intracellular pattern recognition receptor that detects injury signals and initiates inflammatory responses and host defense. Furthermore, NOD1 serves as a metabolic mediator by influencing the metabolism of various tissues, including adipose tissue, liver, cardiovascular tissue, pancreatic β cells, adrenal glands, and bones through diverse mechanisms. It has been discovered that activated NOD1 is associated with the pathological mechanisms of certain metabolic diseases. This review presents a comprehensive summary of the impact of NOD1 on tissue-specific metabolism.
Collapse
Affiliation(s)
- Ruobing Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiyu Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Chengdu, Sichuan, China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Kroh A, Schmitz S, Köhne S, Andruszkow J, Nolting J, Preisinger C, Große K, Eickhoff RM, Heise D, Cramer T, Rheinwalt KP, Alizai PH, Neumann UP, Ulmer TF. Sleeve-gastrectomy results in improved metabolism and a massive stress response of the liver proteome in a mouse model of metabolic dysfunction-associated steatohepatitis. Heliyon 2024; 10:e38678. [PMID: 39524892 PMCID: PMC11550656 DOI: 10.1016/j.heliyon.2024.e38678] [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: 03/12/2024] [Revised: 09/21/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024] Open
Abstract
Background Bariatric surgery has been shown to improve the histopathological findings in patients with obesity and metabolic dysfunction-associated steatohepatitis, but there are also reports about non-responders or progressive disease after bariatric interventions. Therefore, it is of utmost importance to understand the pathophysiological processes in the liver after bariatric surgery. Materials and methods In the present study, 4 weeks old male C57/Bl6 mice were fed a Western Diet to induce metabolic dysfunction-associated steatohepatitis and sleeve-gastrectomy (SG), or sham operation in the pair-fed and ad libitum control group were performed. Mice were observed for two or eight weeks after surgery and metabolic assessment was performed throughout the experiment. Histopathology, flow cytometry and proteomic analyses were conducted to evaluate hepatic inflammation, liver metabolism and affected signaling pathways. Results Weight loss was higher, and metabolism significantly improved after SG. Two weeks after SG major inflammatory and regulatory disturbances in the liver were observed. The proportion of hepatic CD3+NK1.1+ cells were decreased, and proteins involved in apoptosis like Fas, Casp1 and Casp9 or in the acute phase response were upregulated in SG mice. These disturbances decreased in the long-term and we observed an increase of many proteins involved in lipid metabolism eight weeks following SG. Conclusions The rapid weight loss and decrease of hepatic fat after SG lead to a proinflammatory response in the liver in the early phase after surgery, which changes to a more moderate immune response in the long-term. We suggest a preoperative risk stratification and postoperative surveillance depending on the histopathological findings.
Collapse
Affiliation(s)
- Andreas Kroh
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Sophia Schmitz
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Saskia Köhne
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Julia Andruszkow
- Institute of Pathology, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Jochen Nolting
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Christian Preisinger
- Proteomics Facility, Interdisciplinary Center for Clinical Research (IZKF) Aachen, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Karsten Große
- Department of Medicine III, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074 Aachen, Germany
| | - Roman M. Eickhoff
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Daniel Heise
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Thorsten Cramer
- General-, Visceral-, Pediatric and Transplant Surgery, RWTH Aachen University Hospital, Pauwelsstr. 30, 52074, Aachen, Germany
| | - Karl Peter Rheinwalt
- Department for Bariatric and Metabolic Surgery, St. Franziskus Hospital, Cologne, Germany
| | - Patrick Hamid Alizai
- Department of General and Visceral Surgery, Gemeinschaftskrankenhaus Bonn, Bonn, Germany
| | - Ulf Peter Neumann
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| | - Tom Florian Ulmer
- General-, Visceral- and Transplant Surgery, Uniklinik Essen, Hufelandstr. 55, 45147, Essen, Germany
| |
Collapse
|
8
|
Qiu J, Le Y, Liu N, Chen L, Jiang Y, Wang Y, Fan X, Rong X, Yu Z, Li S, Dou X. Nuciferine Alleviates High-Fat Diet- and ApoE -/--Induced Hepatic Steatosis and Ferroptosis in NAFLD Mice via the PPARα Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:24417-24431. [PMID: 39445611 DOI: 10.1021/acs.jafc.4c04929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) causes significant global mortality and healthcare costs with no recommended pharmacological intervention for clinical management. Nuciferine (Nuc) is an alkaloid with aromatic rings, abundantly found in Nelumbo nucifera Gaertn. In this study, we explored the protective mechanisms of Nuc against hepatic steatosis and ferroptosis in NAFLD. High-fat diet (HFD) and healthy male ApoE-/- mice were used to induce NAFLD and a hypercholesterolemia model. Nuc was administered to the mice for four consecutive weeks from the ninth week. Various assessments, including histopathology, RNA sequencing, lipid metabolism, and ferroptosis-related protein expression, showed that Nuc alleviated hepatic steatosis and ferroptosis. We further showed that Nuc improves fatty acid accumulation and ferroptosis through the PPARα signaling pathway in mice and RSL3-treated AML-12 cells. The PPARα inhibitor GW6471 blocked Nuc's protective effects, leading to excess accumulation of iron ions. Thus, Nuc may be a potential therapeutic agent for NAFLD.
Collapse
Affiliation(s)
- Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
- School of Public Health, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Yifei Le
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Nian Liu
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Yuwei Jiang
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Yuhao Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease of The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang University School of Medicine, 268 Kaixuan Road, Shangcheng District, Hangzhou, Zhejiang 310020, China
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, No. 38 Zheda Road, Hangzhou, Zhejiang 310027, China
| | - Xianglu Rong
- Chinese Medicine Institute, Guangdong Pharmaceutical University, No. 280 Outer Ring East Road, University Town, Guangzhou, Guangdong 510006, China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Hong Kong 999077, China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, No. 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang 310053, China
| |
Collapse
|
9
|
Nascimento AL, Pereira JHS, Caldas BV, Guimarães VHD, Monteiro-Junior RS, Paula AMB, Guimarães ALS, Pereira UA, Santos SHS. Dietary Supplementation with Apis mellifera Wholemeal Flour Reduces Hepatic Steatosis in Obese Mice. J Med Food 2024; 27:545-551. [PMID: 38770674 DOI: 10.1089/jmf.2023.0201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Affiliation(s)
- Aline L Nascimento
- Instituto de Ciências Agrárias (ICA), Postgraduate Program in Food and Health, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Brazil
| | - Joyce H S Pereira
- Laboratory of Health Sciences, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Brazil
| | - Bruna V Caldas
- Instituto de Ciências Agrárias (ICA), Postgraduate Program in Food and Health, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Brazil
| | - Victor H D Guimarães
- Laboratory of Health Sciences, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Brazil
| | - Renato S Monteiro-Junior
- Laboratory of Health Sciences, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Brazil
| | - Alfredo M B Paula
- Laboratory of Health Sciences, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Brazil
| | - André L S Guimarães
- Laboratory of Health Sciences, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Brazil
| | - Ulisses A Pereira
- Instituto de Ciências Agrárias (ICA), Postgraduate Program in Food and Health, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Brazil
| | - Sérgio H S Santos
- Instituto de Ciências Agrárias (ICA), Postgraduate Program in Food and Health, Universidade Federal de Minas Gerais (UFMG), Montes Claros, Brazil
- Laboratory of Health Sciences, Postgraduate Program in Health Sciences, Universidade Estadual de Montes Claros (UNIMONTES), Montes Claros, Brazil
| |
Collapse
|
10
|
Jia R, Hou Y, Zhang L, Li B, Zhu J. Effects of Berberine on Lipid Metabolism, Antioxidant Status, and Immune Response in Liver of Tilapia ( Oreochromis niloticus) under a High-Fat Diet Feeding. Antioxidants (Basel) 2024; 13:548. [PMID: 38790653 PMCID: PMC11117941 DOI: 10.3390/antiox13050548] [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: 04/14/2024] [Revised: 04/28/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024] Open
Abstract
Berberine, a natural alkaloid found abundantly in various medicinal plants, exhibits antioxidative, anti-inflammatory, and lipid metabolism-regulatory properties. Nonetheless, its protective effects and the molecular mechanisms underlying liver injury in fish have not been fully elucidated. The aims of this study were to investigate the antioxidative, anti-inflammatory, and lipid metabolism-regulating effects of berberine against high-fat diet (HFD)-induced liver damage and to clarify the underlying molecular mechanisms. Tilapia were fed diets containing two doses of berberine (50 and 100 mg/kg diet) alongside high fat for 60 days. The results showed that berberine treatments (50 and/or 100 mg/kg) significantly reduced elevated aminotransferases, triglycerides (TG), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-c) in the plasma. In the liver, berberine treatments significantly increased the expression of peroxisome proliferator-activated receptor α (pparα) and carnitine palmitoyltransferase 1 (cpt-1) genes, leading to a reduction in lipid accumulation. Meanwhile, berberine treatment suppressed lipid peroxidation formation and enhanced antioxidant capacity. Berberine upregulated the mRNA levels of erythroid 2-related factor 2 (nrf2) and its downstream genes including heme oxygenase 1 (ho-1) and glutathione-S-transferase (gstα). Additionally, berberine attenuated the inflammation by inhibiting the expression of toll-like receptor 2 (tlr2), myeloid differential protein-88 (myd88), relb, and inflammatory cytokines such as interleukin-1β (il-1β), tumor necrosis factor-α (tnf-α), and il-8. In summary, this study suggested that berberine offers protection against HFD-induced liver damage in tilapia via regulating lipid metabolism, antioxidant status, and immune response. This protective effect may be attributed to the modulation of the Nrf2, TLR2/MyD88/NF-κB, and PPARα signaling pathways.
Collapse
Affiliation(s)
- Rui Jia
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (R.J.); (Y.H.); (L.Z.)
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Yiran Hou
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (R.J.); (Y.H.); (L.Z.)
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Liqiang Zhang
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (R.J.); (Y.H.); (L.Z.)
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Bing Li
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (R.J.); (Y.H.); (L.Z.)
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| | - Jian Zhu
- Key Laboratory of Integrated Rice-Fish Farming Ecology, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, Chinese Academy of Fishery Sciences, Wuxi 214081, China; (R.J.); (Y.H.); (L.Z.)
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi 214081, China
| |
Collapse
|
11
|
Li Z, Zheng D, Zhang T, Ruan S, Li N, Yu Y, Peng Y, Wang D. The roles of nuclear receptors in cholesterol metabolism and reverse cholesterol transport in nonalcoholic fatty liver disease. Hepatol Commun 2024; 8:e0343. [PMID: 38099854 PMCID: PMC10727660 DOI: 10.1097/hc9.0000000000000343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023] Open
Abstract
As the most prevalent chronic liver disease globally, NAFLD encompasses a pathological process that ranges from simple steatosis to NASH, fibrosis, cirrhosis, and HCC, closely associated with numerous extrahepatic diseases. While the initial etiology was believed to be hepatocyte injury caused by lipid toxicity from accumulated triglycerides, recent studies suggest that an imbalance of cholesterol homeostasis is of greater significance. The role of nuclear receptors in regulating liver cholesterol homeostasis has been demonstrated to be crucial. This review summarizes the roles and regulatory mechanisms of nuclear receptors in the 3 main aspects of cholesterol production, excretion, and storage in the liver, as well as their cross talk in reverse cholesterol transport. It is hoped that this review will offer new insights and theoretical foundations for the study of the pathogenesis and progression of NAFLD and provide new research directions for extrahepatic diseases associated with NAFLD.
Collapse
|
12
|
Singh P, Kaur L, Ghose S, Varshney S, Jyothi V, Ghosh S, Kommineni P, Kv S, Scaria V, Sivasubbu S, Chandak GR, Sengupta S. Maternal-Periconceptional Vitamin B12 Deficiency in Wistar Rats Leads to Sex-Specific Programming for Cardiometabolic Disease Risk in the Next Generation. J Nutr 2023; 153:3382-3396. [PMID: 37660953 DOI: 10.1016/j.tjnut.2023.08.032] [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: 06/16/2023] [Revised: 08/18/2023] [Accepted: 08/24/2023] [Indexed: 09/05/2023] Open
Abstract
BACKGROUND Maternal vitamin B12 deficiency plays a vital role in fetal programming, as corroborated by previous studies on murine models and longitudinal human cohorts. OBJECTIVES This study assessed the effects of diet-induced maternal vitamin B12 deficiency on F1 offspring in terms of cardiometabolic health and normalization of these effects by maternal-periconceptional vitamin B12 supplementation. METHODS A diet-induced maternal vitamin B12 deficient Wistar rat model was generated in which female rats were either fed a control AIN-76A diet (with 0.01 g/kg vitamin B12) or the same diet with vitamin B12 removed. Females from the vitamin B12-deficient group were mated with males on the control diet. A subset of vitamin B12-deficient females was repleted with vitamin B12 on day 1 of conception. The offspring in the F1 generation were assessed for changes in body composition, plasma biochemistry, and molecular changes in the liver. A multiomics approach was used to obtain a mechanistic insight into the changes in the offspring liver. RESULTS We showed that a 36% reduction in plasma vitamin B12 levels during pregnancy in F0 females can lead to continued vitamin B12 deficiency (60%-70% compared with control) in the F1 offspring and program them for cardiometabolic adversities. These adversities, such as high triglycerides and low high-density lipoprotein cholesterol, were seen only among F1 males but not females. DNA methylome analysis of the liver of F1 3-mo-old offspring highlights sexual dimorphism in the alteration of methylation status of genes critical to signaling processes. Proteomics and targeted metabolomics analysis confirm that sex-specific alterations occur through modulations in PPAR signaling and steroid hormone biosynthesis pathway. Repletion of deficient mothers with vitamin B12 at conception normalizes most of the molecular and biochemical changes. CONCLUSIONS Maternal vitamin B12 deficiency has a programming effect on the next generation and increases the risk for cardiometabolic syndrome in a sex-specific manner. Normalization of the molecular risk markers on vitamin B12 supplementation indicates a causal role.
Collapse
Affiliation(s)
- Praveen Singh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Lovejeet Kaur
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India; Translational Health Science and Technology Institute, Faridabad, India
| | - Subhoshree Ghose
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Swati Varshney
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Vislavath Jyothi
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Sourav Ghosh
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | | | - Shamsudheen Kv
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India
| | - Vinod Scaria
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sridhar Sivasubbu
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Giriraj Ratan Chandak
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| | - Shantanu Sengupta
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
13
|
Zeng QY, Zhang F, Zhang JH, Hei Z, Li ZH, Huang MH, Fang P, Wang ED, Sun XJ, Zhou XL. Loss of threonyl-tRNA synthetase-like protein Tarsl2 has little impact on protein synthesis but affects mouse development. J Biol Chem 2023; 299:104704. [PMID: 37059185 DOI: 10.1016/j.jbc.2023.104704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/16/2023] Open
Abstract
Aminoacyl-tRNA synthetases (aaRSs) are essential components for mRNA translation. Two sets of aaRSs are required for cytoplasmic and mitochondrial translation in vertebrates. Interestingly, TARSL2 is a recently evolved duplicated gene of TARS1 (encoding cytoplasmic threonyl-tRNA synthetase) and represents the only duplicated aaRS gene in vertebrates. Although TARSL2 retains the canonical aminoacylation and editing activities in vitro, whether it is a true tRNA synthetase for mRNA translation in vivo is unclear. In this study, we showed that Tars1 is an essential gene since homozygous Tars1 knockout mice were lethal. In contrast, when Tarsl2 was deleted in mice and zebrafish, neither the abundance nor the charging levels of tRNAThrs were changed, indicating that cells relied on Tars1 but not on Tarsl2 for mRNA translation. Furthermore, Tarsl2 deletion did not influence the integrity of the multiple tRNA synthetase complex (MSC), suggesting that Tarsl2 is a peripheral member of the MSC. Finally, we observed that Tarsl2-deleted mice exhibited severe developmental retardation, elevated metabolic capacity, and abnormal bone and muscle development after 3 weeks. Collectively, these data suggest that, despite its intrinsic activity, loss of Tarsl2 has little influence on protein synthesis but does affect mouse development.
Collapse
Affiliation(s)
- Qi-Yu Zeng
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031
| | - Fan Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200010
| | - Jian-Hui Zhang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024
| | - Zhoufei Hei
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China; School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zi-Han Li
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031
| | - Meng-Han Huang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031
| | - Pengfei Fang
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai 200032, China; School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| | - En-Duo Wang
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031.
| | - Xiao-Jian Sun
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200010.
| | - Xiao-Long Zhou
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031; School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024.
| |
Collapse
|
14
|
Junli Z, Shuhan W, Yajuan Z, Xiaoling D, Jiahuan L, Keshu X. The Role and Mechanism of CREBH Regulating SIRT3 in Metabolic Associated Fatty Liver Disease. Life Sci 2022; 306:120838. [PMID: 35902030 DOI: 10.1016/j.lfs.2022.120838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/04/2022] [Accepted: 07/20/2022] [Indexed: 10/16/2022]
Abstract
AIMS To investigate the effect of cAMP response element-binding protein H (CREBH) on metabolic associated fatty liver disease by regulating sirtuin 3 (SIRT3). MAIN METHODS Two mouse models of fatty liver induced by a methionine-choline deficient (MCD) diet and a high-fat (HF) diet and an in vitro model of palmitic acid (PA) induced lipid-overloaded hepatocytes were constructed to detect the expression of CREBH, SIRT3, total acetylation, and downstream protein interactions and lipid metabolism phenotype, which were further validated in CREBH-/- mice and lentivirus-overexpressing CREBH hepatocytes. KEY FINDINGS In fatty liver and lipid overload models, the expressions of CREBH and SIRT3 were down-regulated and their expression was positively correlated, accompanied by an increase in the level of total protein acetylation. Overexpression of CREBH alleviated excess lipid accumulation, impaired viability, and the ability to metabolize energy through the fatty acid oxidation pathway in hepatocytes in vitro. Furthermore, overexpression of CREBH restored the interaction of the deacetylase SIRT3 with the molecules carnitine palmitoyl-transferase 2 (CPT2) and long-chain acyl CoA dehydrogenase (ACADL) involved in the fatty acid oxidation pathway and their deacetylation status. However, CREBH-/- aggravated the damage of lipid metabolism in the liver tissue of mice. SIGNIFICANCE CREBH increased the enzymatic activity of downstream factors by positively regulating the expression of SIRT3, which promoted the oxidative decomposition of fatty acids in hepatocytes and played an important role in fatty acid oxidation in MAFLD.
Collapse
Affiliation(s)
- Zhang Junli
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wang Shuhan
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhao Yajuan
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Deng Xiaoling
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Li Jiahuan
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xu Keshu
- Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
15
|
Effects of Stocking Density on the Growth Performance, Physiological Parameters, Redox Status and Lipid Metabolism of Micropterus salmoides in Integrated Rice–Fish Farming Systems. Antioxidants (Basel) 2022; 11:antiox11071215. [PMID: 35883706 PMCID: PMC9312047 DOI: 10.3390/antiox11071215] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/14/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022] Open
Abstract
Stocking density has been identified as one of the main factors affecting fish growth, welfare and behavior. However, few studies have focused on the effects of stocking density on fish health in integrated rice–fish farming systems. Thus, the aim of this study was to evaluate the effects of different stocking densities on the growth performance, physiological parameters, redox status and lipid metabolism of Micropterus salmoides in an integrated rice–fish farming system. The fish were reared at three densities: low density (LD, 40 g/m3), medium density (MD, 80 g/m3) and high density (HD, 120 g/m3) for 90 days. At the end of the experiment, fish reared in the MD and HD groups showed lower growth performance than those from the LD group. The HD treatment significantly altered the physiological parameters, including glucose and lactate. Meanwhile, the HD treatment induced oxidative stress and lipid peroxidation after 90 days of farming. Furthermore, transcriptomic analysis revealed that HD treatment led to abnormal lipid metabolism. Interestingly, we found the suppression of three key pathways related to lipid metabolism, including the PPAR, insulin and adipocytokine signaling pathways, in the HD group. Overall, our data indicated that the HD treatment inhibited growth and caused physiological responses, oxidative stress and abnormal hepatic lipid metabolism in M. salmoides in an integrated rice–fish farming system.
Collapse
|
16
|
Abot A, Brochot A, Pomié N, Wemelle E, Druart C, Régnier M, Delzenne NM, de Vos WM, Knauf C, Cani PD. Camu-Camu Reduces Obesity and Improves Diabetic Profiles of Obese and Diabetic Mice: A Dose-Ranging Study. Metabolites 2022; 12:metabo12040301. [PMID: 35448490 PMCID: PMC9025096 DOI: 10.3390/metabo12040301] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/21/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
Overweight, obesity, and their comorbidities are currently considered a major public health concern. Today considerable efforts are still needed to develop efficient strategies able to attenuate the burden of these diseases. Nutritional interventions, some with plant extracts, present promising health benefits. In this study, we evaluated the action of Camu-Camu (Myrciaria dubia), an Amazonian fruit rich in polyphenols and vitamin C, on the prevention of obesity and associated disorders in mice and the abundance of Akkermansia muciniphila in both cecum and feces. Methods: We investigated the dose-response effects of Camu-Camu extract (CCE) in the context of high-fat-diet (HFD)-induced obesity. After 5 weeks of supplementation, we demonstrated that the two doses of CCE differently improved glucose and lipid homeostasis. The lowest CCE dose (62.5 mg/kg) preferentially decreased non-HDL cholesterol and free fatty acids (FFA) and increased the abundance of A. muciniphila without affecting liver metabolism, while only the highest dose of CCE (200 mg/kg) prevented excessive body weight gain, fat mass gain, and hepatic steatosis. Both doses decreased fasting hyperglycemia induced by HFD. In conclusion, the use of plant extracts, and particularly CCE, may represent an additional option in the support of weight management strategies and glucose homeostasis alteration by mechanisms likely independent from the modulation of A. muciniphila abundance.
Collapse
Affiliation(s)
- Anne Abot
- Enterosys SAS, 31670 Labège, France; (A.A.); (N.P.)
| | - Amandine Brochot
- A-Mansia Biotech SA, The Akkermansia Company, 1435 Mont-Saint-Guibert, Belgium; (A.B.); (C.D.)
| | | | - Eve Wemelle
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Université Paul Sabatier (UPS), 31000 Toulouse, France;
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
| | - Céline Druart
- A-Mansia Biotech SA, The Akkermansia Company, 1435 Mont-Saint-Guibert, Belgium; (A.B.); (C.D.)
| | - Marion Régnier
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
| | - Nathalie M. Delzenne
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Claude Knauf
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Institut de Recherche en Santé Digestive et Nutrition (IRSD), Université Paul Sabatier (UPS), 31000 Toulouse, France;
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
- Correspondence: (C.K.); (P.D.C.)
| | - Patrice D. Cani
- NeuroMicrobiota Lab, International Research Program (IRP) INSERM, 31000 Toulouse, France
- WELBIO—Walloon Excellence in Life Sciences and BIOtechnology, Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium; (M.R.); (N.M.D.)
- Correspondence: (C.K.); (P.D.C.)
| |
Collapse
|
17
|
Yagai T, Nakamura T. Mechanistic insights into the peroxisome proliferator-activated receptor alpha as a transcriptional suppressor. Front Med (Lausanne) 2022; 9:1060244. [PMID: 36507526 PMCID: PMC9732035 DOI: 10.3389/fmed.2022.1060244] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/08/2022] [Indexed: 11/27/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most prevalent hepatic disorders that 20-30% of the world population suffers from. The feature of NAFLD is excess lipid accumulation in the liver, exacerbating multiple metabolic syndromes such as hyperlipidemia, hypercholesterolemia, hypertension, and type 2 diabetes. Approximately 20-30% of NAFLD cases progress to more severe chronic hepatitis, known as non-alcoholic steatohepatitis (NASH), showing deterioration of hepatic functions and liver fibrosis followed by cirrhosis and cancer. Previous studies uncovered that several metabolic regulators had roles in disease progression as key factors. Peroxisome proliferator-activated receptor alpha (PPARα) has been identified as one of the main players in hepatic lipid homeostasis. PPARα is abundantly expressed in hepatocytes, and is a ligand-dependent nuclear receptor belonging to the NR1C nuclear receptor subfamily, orchestrating lipid/glucose metabolism, inflammation, cell proliferation, and carcinogenesis. PPARα agonists are expected to be novel prescription drugs for NASH treatment, and some of them (e.g., Lanifibranor) are currently under clinical trials. These potential novel drugs are developed based on the knowledge of PPARα-activating target genes related to NAFLD and NASH. Intriguingly, PPARα is known to suppress the expression of subsets of target genes under agonist treatment; however, the mechanisms of PPARα-mediated gene suppression and functions of these genes are not well understood. In this review, we summarize and discuss the mechanisms of target gene repression by PPARα and the roles of repressed target genes on hepatic lipid metabolism, fibrosis and carcinogenesis related to NALFD and NASH, and provide future perspectives for PPARα pharmaceutical potentials.
Collapse
Affiliation(s)
- Tomoki Yagai
- Department of Metabolic Bioregulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Takahisa Nakamura
- Department of Metabolic Bioregulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.,Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
18
|
Quiroga S, Juárez YR, Marcone MP, Vidal MA, Genaro AM, Burgueño AL. Prenatal stress promotes insulin resistance without inflammation or obesity in C57BL/6J male mice. Stress 2021; 24:987-997. [PMID: 34581257 DOI: 10.1080/10253890.2021.1978425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
During gestation, stress exposure increases the risk of developing cognitive and physiological alterations in either the long or short term. Among them, metabolic alterations have been described. Adipose tissue is responsible for the secretion of several factors involved in controlling body weight and energy expenditure, the regulation of insulin sensitivity, and the development of inflammation, among others. Moreover, the liver regulates glucose homeostasis and lipid metabolism, playing an essential role in developing insulin resistance. In this work, we analyzed if prenatal stress leads to alterations in metabolism and the relationship between these alterations and gene expression in the adipose tissue and the liver. Prenatal stress-exposed animals developed disturbances in the glucose and insulin response curve, showing in both tests higher glycemia than the control group. However, they did not exhibit increased body weight. At the same time, in the adipose tissue, we observed an increase in mRNA expression of Leptin and Resistin and a decrease in Adiponectin. In the liver, we observed a lower mRNA expression of several genes involved in glucose metabolism and fatty acid oxidation, such as Sirt1, Pgc1α, Pparα, among others. In both tissues, we observed a lower expression of inflammatory genes. These results suggest that prenatal stress exposure produces insulin resistance at both physiological and molecular levels without pro-inflammatory signaling or obesity.
Collapse
Affiliation(s)
- Sofia Quiroga
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Yamila Raquel Juárez
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - María Paula Marcone
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - María Agustina Vidal
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Ana María Genaro
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| | - Adriana Laura Burgueño
- Instituto de Investigaciones Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Pontificia Universidad Católica Argentina, Buenos Aires, Argentina
| |
Collapse
|
19
|
Transcriptome analysis to reveal the mechanism of the effect of Echinops latifolius polysaccharide B on palmitate-induced insulin-resistant. Biomed Pharmacother 2021; 143:112203. [PMID: 34563954 DOI: 10.1016/j.biopha.2021.112203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/08/2021] [Accepted: 09/13/2021] [Indexed: 01/13/2023] Open
Abstract
Hepatic insulin resistance is a crucial pathological process in type 2 diabetes mellitus (T2DM) associated with visceral adiposity and metabolic disorders. Echinops latifolius polysaccharide B (ETPB), a polysaccharide extracted from Echinops latifolius Tausch, increases insulin sensitivity in the high-fat diet-fed and STZ induced SD rat model and even prevented hepatic metabolic disorders. However, the mechanism by which ETPB improves carbohydrate and lipid metabolisms in the liver with insulin resistance remains largely unknown. In the present work, an lnsulin resistance cell model (IR-HepG2) was established. Glucose consumption, glycogen content, triglycerides (TG), and free fatty acids (FFAs) levels were detected. The result revealed that the intervention of ETPB significantly increased glucose consumption and glycogen synthesis and reduced FFAs and TG production in IR-HepG2 cells. Further, we also employed RNA-seq to identify differentially expressed miRNAs (DEMs) and mRNAs (DEGs) with a fold change of ≥ 1.5 and p-value of <0.05. Finally, we identified 1028, 682, 382, 1614, 519 and 825 DEGs, and 71, 113, 94, 68, 52 and 38 DEMs in different comparisons, respectively. Based on a short time-series expression miner (STEM) analysis, six profiles were chosen for further analysis. Seventeen insulin resistance-associated dynamic DEGs were identified during ETPB stimulation. Based on these dynamic DEGs, the related miRNAs were acquired from DEMs, and an integrated miRNA-mRNA regulatory network was subsequently constructed. Besides, some DEGs and DEMs were validated using qPCR. This study provides transcriptomic evidence of the molecular mechanism involved in HepG2 insulin resistance, leading to the discovery of miRNA-based target therapies for ETPB.
Collapse
|
20
|
Epithelial Cell Transformation and Senescence as Indicators of Genome Aging: Current Advances and Unanswered Questions. Int J Mol Sci 2021; 22:ijms22147544. [PMID: 34299168 PMCID: PMC8303760 DOI: 10.3390/ijms22147544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 02/06/2023] Open
Abstract
The recent advances in deciphering the human genome allow us to understand and evaluate the mechanisms of human genome age-associated transformations, which are largely unclear. Genome sequencing techniques assure comprehensive mapping of human genetics; however, understanding of gene functional interactions, specifically of time/age-dependent modifications, remain challenging. The age of the genome is defined by the sum of individual (inherited) and acquired genomic traits, based on internal and external factors that impact ontogenesis from the moment of egg fertilization and embryonic development. The biological part of genomic age opens a new perspective for intervention. The discovery of single cell-based mechanisms for genetic change indicates the possibility of influencing aging and associated disease burden, as well as metabolism. Cell populations with transformed genetic background were shown to serve as the origin of common diseases during extended life expectancy (superaging). Consequently, age-related cell transformation leads to cancer and cell degeneration (senescence). This article aims to describe current advances in the genomic mechanisms of senescence and its role in the spatiotemporal spread of epithelial clones and cell evolution.
Collapse
|
21
|
Baiocchi L, Lenci I, Milana M, Kennedy L, Sato K, Zhang W, Ekser B, Ceci L, Meadows V, Glaser S, Alpini G, Francis H. Cyclic AMP Signaling in Biliary Proliferation: A Possible Target for Cholangiocarcinoma Treatment? Cells 2021; 10:1692. [PMID: 34359861 PMCID: PMC8303798 DOI: 10.3390/cells10071692] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Cholangiocarcinoma is a lethal disease with scarce response to current systemic therapy. The rare occurrence and large heterogeneity of this cancer, together with poor knowledge of its molecular mechanisms, are elements contributing to the difficulties in finding an appropriate cure. Cholangiocytes (and their cellular precursors) are considered the liver component giving rise to cholangiocarcinoma. These cells respond to several hormones, neuropeptides and molecular stimuli employing the cAMP/PKA system for the translation of messages in the intracellular space. For instance, in physiological conditions, stimulation of the secretin receptor determines an increase of intracellular levels of cAMP, thus activating a series of molecular events, finally determining in bicarbonate-enriched choleresis. However, activation of the same receptor during cholangiocytes' injury promotes cellular growth again, using cAMP as the second messenger. Since several scientific pieces of evidence link cAMP signaling system to cholangiocytes' proliferation, the possible changes of this pathway during cancer growth also seem relevant. In this review, we summarize the current findings regarding the cAMP pathway and its role in biliary normal and neoplastic cell proliferation. Perspectives for targeting the cAMP machinery in cholangiocarcinoma therapy are also discussed.
Collapse
Affiliation(s)
- Leonardo Baiocchi
- Hepatology Unit, University of Tor Vergata, 00133 Rome, Italy; (L.B.); (I.L.); (M.M.)
| | - Ilaria Lenci
- Hepatology Unit, University of Tor Vergata, 00133 Rome, Italy; (L.B.); (I.L.); (M.M.)
| | - Martina Milana
- Hepatology Unit, University of Tor Vergata, 00133 Rome, Italy; (L.B.); (I.L.); (M.M.)
| | - Lindsey Kennedy
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Keisaku Sato
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
| | - Wenjun Zhang
- Division of Transplant Surgery, Department of Surgery, Indiana University, Indianapolis, IN 46202, USA; (W.Z.); (B.E.)
| | - Burcin Ekser
- Division of Transplant Surgery, Department of Surgery, Indiana University, Indianapolis, IN 46202, USA; (W.Z.); (B.E.)
| | - Ludovica Ceci
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
| | - Vik Meadows
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
| | - Shannon Glaser
- Department of Medical Physiology, Texas A&M University College of Medicine, Bryan, TX 77807, USA;
| | - Gianfranco Alpini
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| | - Heather Francis
- Hepatology and Gastroenterology, Medicine, Indiana University, Indianapolis, IN 46202, USA; (L.K.); (K.S.); (L.C.); (V.M.)
- Richard L. Roudebush VA Medical Center, Indianapolis, IN 46202, USA
| |
Collapse
|