1
|
Li Q, Sheng J, Baruscotti M, Liu Z, Wang Y, Zhao L. Identification of Senkyunolide I as a novel modulator of hepatic steatosis and PPARα signaling in zebrafish and hamster models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118743. [PMID: 39209000 DOI: 10.1016/j.jep.2024.118743] [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: 07/01/2024] [Revised: 08/19/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Non-alcoholic fatty liver disease (NAFLD) is the leading cause of liver-related morbidity and mortality, with hepatic steatosis being the hallmark symptom. Salvia miltiorrhiza Bunge (Smil, Dan-Shen) and Ligusticum striatum DC (Lstr, Chuan-Xiong) are commonly used to treat cardiovascular diseases and have the potential to regulate lipid metabolism. However, whether Smil/Lstr combo can be used to treat NAFLD and the mechanisms underlying its lipid-regulating properties remain unclear. PURPOSE To assess the feasibility and reliability of a short-term high-fat diet (HFD) induced zebrafish model for evaluating hepatic steatosis phenotype and to investigate the liver lipid-lowering effects of Smil/Lstr, as well as its active components. METHODS The phenotypic alterations of liver and multiple other organ systems were examined in the HFD zebrafish model using fluorescence imaging and histochemistry. The liver-specific lipid-lowering effects of Smil/Lstr combo were evaluated endogenously. The active molecules and functional mechanisms were further explored in zebrafish, human hepatocytes, and hamster models. RESULTS In 5-day HFD zebrafish, significant lipid accumulation was detected in the blood vessels and the liver, as evidenced by increased staining with Oil Red O and fluorescent lipid probes. Hepatic hypertrophy was observed in the model, along with macrovesicular steatosis. Smil/Lstr combo administration effectively restored the lipid profile and alleviated hepatic hypertrophy in the HFD zebrafish. In oleic-acid stimulated hepatocytes, Smil/Lstr combo markedly reduced lipid accumulation and cell damage. Subsequently, based on zebrafish phenotypic screening, the natural phthalide senkyunolide I (SEI) was identified as a major molecule mediating the lipid-lowering activities of Smil/Lstr combo in the liver. Moreover, SEI upregulated the expression of the lipid metabolism regulator PPARα and downregulated fatty acid translocase CD36, while a PPARα antagonist sufficiently blocked the regulatory effect of SEI on hepatic steatosis. Finally, the roles of SEI on hepatic lipid accumulation and PPARα signaling were further verified in the hamster model. CONCLUSIONS We proposed a zebrafish-based screening strategy for modulators of hepatic steatosis and discovered the regulatory roles of Smil/Lstr combo and its component SEI on liver lipid accumulation and PPARα signaling, suggesting their potential value as novel candidates for NAFLD treatment.
Collapse
Affiliation(s)
- Qingquan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jian Sheng
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Mirko Baruscotti
- Department of Biosciences, University of Milano, Milan, 1-20133, Italy
| | - Zhenjie Liu
- Department of Vascular Surgery, The Second Affiliated Hospital of Zhejiang University Medical School, Hangzhou, 310003, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310020, China
| | - Lu Zhao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China; Department of Vascular Surgery, The Second Affiliated Hospital of Zhejiang University Medical School, Hangzhou, 310003, China; State Key Laboratory of Chinese Medicine Modernization, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Al Harake SN, Abedin Y, Hatoum F, Nassar NZ, Ali A, Nassar A, Kanaan A, Bazzi S, Azar S, Harb F, Ghadieh HE. Involvement of a battery of investigated genes in lipid droplet pathophysiology and associated comorbidities. Adipocyte 2024; 13:2403380. [PMID: 39329369 PMCID: PMC11445895 DOI: 10.1080/21623945.2024.2403380] [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: 12/07/2023] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LDs) are highly specialized energy storage organelles involved in the maintenance of lipid homoeostasis by regulating lipid flux within white adipose tissue (WAT). The physiological function of adipocytes and LDs can be compromised by mutations in several genes, leading to NEFA-induced lipotoxicity, which ultimately manifests as metabolic complications, predominantly in the form of dyslipidemia, ectopic fat accumulation, and insulin resistance. In this review, we delineate the effects of mutations and deficiencies in genes - CIDEC, PPARG, BSCL2, AGPAT2, PLIN1, LIPE, LMNA, CAV1, CEACAM1, and INSR - involved in lipid droplet metabolism and their associated pathophysiological impairments, highlighting their roles in the development of lipodystrophies and metabolic dysfunction.
Collapse
Affiliation(s)
- Sami N. Al Harake
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Yasamin Abedin
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Fatema Hatoum
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Nour Zahraa Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Ali Ali
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Aline Nassar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Amjad Kanaan
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Samer Bazzi
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Sami Azar
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Frederic Harb
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| | - Hilda E. Ghadieh
- Department of Biomedical Sciences, Faculty of Medicine and Medical Sciences, University of Balamand, Kalhat, Lebanon
| |
Collapse
|
3
|
Li YN, Sun FF, Ouyang F, Luo D, Zhang ZX, Lu MX, Hu CY, Shi YH, Gui Q, Zhang JY, Yang TS. Alterations in liver triglyceride profiles in CCl 4-induced liver regeneration. Biochem Biophys Res Commun 2024; 734:150662. [PMID: 39245030 DOI: 10.1016/j.bbrc.2024.150662] [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: 08/21/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/10/2024]
Abstract
Lipid metabolism, particularly triglyceride (TG) metabolism, is crucial for liver regeneration. During the early phase of liver regeneration, the liver temporarily accumulates a substantial amount of TG-dominated lipids. However, the specific composition of the TG profile during this phase is not yet fully understood. Here, we showed that the TG molecular composition in the liver was significantly altered during liver regeneration following carbon tetrachloride (CCl4)-induced liver injury. Lipid accumulation in livers was observed as early as 12 hours after CCl4 treatment, with transient regeneration-associated steatosis (TRAS) lasting until 24 hours. Hepatocyte proliferation began only after liver lipid levels returned to baseline at 48 hours. Furthermore, the profile of TG species changed significantly during liver regeneration. During the TRAS period, the accumulated TGs in the liver were mainly long-chain triglycerides, with most of the fatty acids constituting these triglycerides having fewer than 20 carbon atoms. In the proliferation phase, the fatty acid composition of these triglycerides shifted from long-chain to ultra-long-chain fatty acids. Our results suggest a significant TRAS-related change in the TG lipid profile of the liver during liver regeneration.
Collapse
Affiliation(s)
- Yi-Ning Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Fang-Fang Sun
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Feng Ouyang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Da Luo
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Zi-Xuan Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Man-Xi Lu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Chang-Yong Hu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Yi-Hong Shi
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Qian Gui
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Jia-Yi Zhang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Tian-Shu Yang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Huang Y, Xin H, Lin Q, Yang G, Zhang Y, Cao D, Yu X. A fluorescent probe for detecting bisulfite/sulfite in lipid droplets and tracking the dynamics of lipid droplets. Talanta 2024; 279:126605. [PMID: 39084038 DOI: 10.1016/j.talanta.2024.126605] [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: 04/19/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024]
Abstract
Intracellular lipid droplets (LDs) are important organelles regulating intracellular redox processes. Endogenous bisulfite/sulfite (HSO3-/SO32-) is one of the metabolites of thiol metabolism. The variation in HSO3-/SO32- content around LDs is closely related to cellular homeostasis. However, there is currently no effective method to visualize and quantify the dynamic changes in HSO3-/SO32- content around LDs. In this work, a fluorescent probe MC-BEN utilizing a triphenylamine basic framework was developed to selectively recognize HSO3-/SO32- via a nucleophilic addition reaction. The probe exhibits excellent anti-interference capability, short response time, outstanding photostability, and a low fluorescence detection limit (6.1 μM) for HSO3-/SO32- recognition. More interesting, there is a trend of accelerated contact between LDs and lysosomes after MC-BEN targeting LDs and reacting with endogenous/exogenous HSO3-/SO32-, which may provide new ideas for the study of intracellular lysosomal lipophagy.
Collapse
Affiliation(s)
- Yan Huang
- School of Materials Science and Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Haotian Xin
- School of Materials Science and Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Qiaowen Lin
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, Shandong, China
| | - Guiyi Yang
- School of Materials Science and Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Yan Zhang
- School of Materials Science and Engineering, University of Jinan, Jinan, 250022, Shandong, China
| | - Duxia Cao
- School of Materials Science and Engineering, University of Jinan, Jinan, 250022, Shandong, China.
| | - Xiaoqiang Yu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, Shandong, China.
| |
Collapse
|
5
|
Zhai T, Cai L, Jia X, Xia M, Bian H, Gao X, Pan C, Li X, Xia P. IGFBP2 functions as an endogenous protector against hepatic steatosis via suppression of the EGFR-STAT3 pathway. Mol Metab 2024; 89:102026. [PMID: 39299533 PMCID: PMC11474195 DOI: 10.1016/j.molmet.2024.102026] [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: 06/07/2024] [Revised: 08/17/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is deemed as an emerging global epidemic, whereas the underlying pathogenic mechanism remains to be clarified. We aimed to systemically analyze all the NAFLD-related gene expression datasets from published human-based studies, by which exploring potential key factors and mechanisms accounting for the pathogenesis of NAFLD. METHODS Robust rank aggregation (RRA) method was used to integrate NAFLD-related gene expression datasets. For fatty liver study, adeno-associated virus (AAV) delivery and genetic knockout mice were used to create IGFBP2 (Insulin-like growth factor binding protein 2) gain- or loss-of function models. Western blot, Co-immunoprecipitation (Co-IP), immunofluorescent (IF) staining, luciferase assay, molecular docking simulation were performed to reveal the IGFBP2-EGFR-STAT3 axis involved. Key axis protein levels in livers from healthy donors and patients with NAFLD were assessed via immunohistochemical staining. RESULTS By using RRA method, the present study identified IGFBP2 being the most significantly down-regulated gene in all NAFLD subjects. The decreased IGFBP2 expression was further confirmed in the liver tissues from patients and animal models of NAFLD. IGFBP2 deficiency aggravated hepatic steatosis and NASH phenotypes and promoted lipogenic gene expression both in vivo and in vitro. Mechanistically, IGFBP2 directly binds to and regulates EGFR, whereas blockage of the IGFBP2-EGFR complex by knockdown of IGFBP2 resulted in the EGFR-STAT3 pathway activation, which in turn promoted the promoter activity of Srebf1. By using molecular docking simulation and protein-protein interaction analysis, the sequence of 233-257 amino acids in IGFBP2 was characterized as a key motif responding for its specific binding to EGFR and the protective effect against hepatic steatosis. CONCLUSIONS The current study has, for the first time, identified IGFBP2 as a novel protector against hepatosteatosis. The protective effect is mediated by its specific interaction with EGFR and thereby suppressing the EGFR-STAT3 pathway. Therefore, pharmaceutically targeting the IGFBP2-EGFR-STAT3 axis may provide a theoretical basis for for the treatment of NAFLD/NASH and the associated diseases.
Collapse
Affiliation(s)
- Tianyu Zhai
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Liang Cai
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, China; National Clinical Research Center for Interventional Medicine (NCRC-IM), China; Institute of Vascular Surgery, Fudan University, Shanghai, China.
| | - Xi Jia
- Department of Medicine, School of Clinical Medicine, The University of Hong Kong, Hong Kong S.A.R, China.
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Hua Bian
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Chenling Pan
- Beijing Computing Center, Beijing Academy of Science and Technology, Beijing, China.
| | - Xiaoying Li
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| | - Pu Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, and Fudan Institute for Metabolic Diseases, Fudan University, Shanghai, China.
| |
Collapse
|
6
|
Sun J, Du B, Chen M, Jia J, Wang X, Hong J. FBXO28 reduces high-fat diet-induced hyperlipidemia in mice by alleviating abnormal lipid metabolism and inflammatory responses. J Endocrinol Invest 2024; 47:2757-2774. [PMID: 38696123 DOI: 10.1007/s40618-024-02376-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 04/12/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Hyperlipidemia is a lipid metabolism disorder with increasing incidence and prevalence worldwide. Abnormal lipid metabolism and inflammation are two significant characteristics of hyperlipidemia. The purpose of this study was to explore the role and mechanism of F-box only protein 28 (FBXO28) in hyperlipidemia. METHODS Mice were fed with high-fat diet (HFD) to elicit obesity, and 3T3-L1 preadipocytes were stimulated with MDI cocktail (IBMX, DEX and insulin) to evoke differentiation. In vivo and in vitro role of FBXO28 in hyperlipidemia was investigated by hematoxylin-eosin and oil Red O staining, the lipid biochemistry measurement, enzyme-linked immunosorbent assay, reverse transcription quantitative polymerase chain reaction and western blotting assays. The mechanism of FBXO28 explored by co-immunoprecipitation, immunofluorescence, ubiquitination and cycloheximide assays. RESULTS Low expression of FBXO28 was found in hyperlipidemia in silico, in vivo and in vitro. Upregulation of FBXO28 declined the body weight, fat accumulation, and serum lipid content in HFD-fed mice. Abnormal lipid accumulation, and the level of liposynthetic genes and beta-oxidation related genes were improved by overexpression of FBXO28 both in HFD-elicited mice and MDI-treated 3T3-L1 preadipocytes. Besides, overexpression of FBXO28 declined HFD-induced the level of proinflammatory factors and F4/80. Mechanically, FBXO28 directly bound RAB27A and promoted its ubiquitinated degradation. Thus, upregulation of RAB27A inverted the improved role of FBXO28 in abnormal lipid metabolism and inflammation in vivo and in vitro. CONCLUSION FBXO28 ameliorated abnormal lipid metabolism and inflammation through the ubiquitinated degradation of RAB27A, thereby attenuating HFD-induced hyperlipidemia. The results could promote the treatment of hyperlipidemia, and the relevant diseases.
Collapse
Affiliation(s)
- J Sun
- Cadre's Ward, The Fourth Clinical College of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - B Du
- Department of Neurology, Urumqi Midong District Hospital of Traditional Chinese Medicine, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - M Chen
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - J Jia
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - X Wang
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - J Hong
- Cadre's Ward, Traditional Chinese Medicine Hospital of Xinjiang Medical University, No.116, Huanghe Road, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
7
|
Szkudelski T, Szkudelska K. The relevance of the heme oxygenase system in alleviating diabetes-related hormonal and metabolic disorders. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167552. [PMID: 39490940 DOI: 10.1016/j.bbadis.2024.167552] [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: 09/13/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Heme oxygenase (HO) is an enzyme that catalyzes heme degradation. HO dysfunction is linked to various pathological conditions, including diabetes. Results of animal studies indicate that HO expression and activity are downregulated in experimentally induced diabetes. This is associated with severe hormonal and metabolic disturbances. However, these pathological changes have been shown to be reversed by therapy with HO activators. In animals with experimentally induced diabetes, HO was upregulated by genetic manipulation or by pharmacological activators such as hemin and cobalt protoporphyrin. Induction of HO alleviated elevated blood glucose levels and improved insulin action, among other effects. This effect resulted from beneficial changes in the main insulin-sensitive tissues, i.e., the skeletal muscle, the liver, and the adipose tissue. The action of HO activators was due to positive alterations in pivotal signaling molecules and regulatory enzymes. Furthermore, diabetes-related oxidative and inflammatory stress was reduced due to HO induction. HO upregulation was effective in various animal models of type 1 and type 2 diabetes. These data suggest the possibility of testing HO activators as a potential tool for alleviating hormonal and metabolic disorders in people with diabetes.
Collapse
Affiliation(s)
- Tomasz Szkudelski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35, 60-637 Poznan, Poland.
| | - Katarzyna Szkudelska
- Department of Animal Physiology, Biochemistry and Biostructure, Poznan University of Life Sciences, Wolynska 35, 60-637 Poznan, Poland.
| |
Collapse
|
8
|
Gu B, Jiang Y, Huang Z, Li H, Yu W, Li T, Liu C, Wang P, Chen J, Sun L, Tan P, Fu W, Wen J. MRG15 aggravates sepsis-related liver injury by promoting PCSK9 synthesis and secretion. Int Immunopharmacol 2024; 140:112898. [PMID: 39128417 DOI: 10.1016/j.intimp.2024.112898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
OBJECTIVE Disorders of lipid oxidation play an important role in organ damage, and lipid metabolites are associated with inflammation and coagulation dysfunction in sepsis. However, the specific molecular mechanism by which lipid metabolism-related proteins regulate sepsis is still unclear. The aim of this study is to investigate the role of mortality factor 4-like protein 1 (MORF4L1, also called MRG15), a hepatic lipid metabolism related gene, in sepsis-induced liver injury. METHODS In the mouse sepsis models established by cecal ligation and puncture (CLP) and lipopolysaccharide (LPS), the impact of pretreatment with the MRG15 inhibitor argatroban on sepsis-related liver injury was investigated. In the LPS-induced hepatocyte sepsis cell model, the effects of MRG15 overexpression or knockdown on hepatic inflammation and lipid metabolism were studied. Additionally, in a co-culture system of hepatocytes and macrophages, the influence of MRG15 knockdown in hepatocytes on the synthesis and secretion of inflammation-related protein PCSK9 as well as its effect on macrophage activation were examined. RESULTS Studies have shown that MRG15 expression was increased in septicemia mice and positively correlated with lipid metabolism and inflammation. However, knockdown of MRG15 ameliorates sepsis-induced hepatocyte injury. Increased MRG15 in LPS-stimulated hepatocytes promotes PCSK9 synthesis and secretion, which induces macrophage M1 polarization and exacerbates the inflammatory response. Agatroban, an inhibitor of MRG15, ameliorates sepsis-induced liver injury in mice by inhibiting MRG15-induced lipid metabolism disorders and inflammatory responses. CONCLUSIONS In sepsis, increased MRG15 expression in hepatocytes leads to disturbed hepatic lipid metabolism and induces macrophage M1 polarization by secreting PCSK9, ultimately exacerbating liver injury.
Collapse
Affiliation(s)
- Boyuan Gu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yu Jiang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Zhiwei Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Han Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Wenhao Yu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Tongxi Li
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Pengru Wang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jiatong Chen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Lei Sun
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Peng Tan
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China.
| | - Jian Wen
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China; Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China.
| |
Collapse
|
9
|
Wang H, Nikain C, Fortounas KI, Amengual J, Tufanli O, La Forest M, Yu Y, Wang MC, Watts R, Lehner R, Qiu Y, Cai M, Kurland IJ, Goldberg IJ, Rajan S, Hussain MM, Brodsky JL, Fisher EA. FITM2 deficiency results in ER lipid accumulation, ER stress, and reduced apolipoprotein B lipidation and VLDL triglyceride secretion in vitro and in mouse liver. Mol Metab 2024:102048. [PMID: 39426520 DOI: 10.1016/j.molmet.2024.102048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024] Open
Abstract
OBJECTIVES Triglycerides (TGs) associate with apolipoprotein B100 (apoB100) to form very low density lipoproteins (VLDLs) in the liver. The repertoire of factors that facilitate this association is incompletely understood. FITM2, an integral endoplasmic reticulum (ER) protein, was originally discovered as a factor participating in cytosolic lipid droplet (LD) biogenesis in tissues that do not form VLDL. We hypothesized that in the liver, in addition to promoting cytosolic LD formation, FITM2 would also transfer TG from its site of synthesis in the ER membrane to nascent VLDL particles within the ER lumen. METHODS Experiments were conducted using a rat hepatic cell line (McArdle-RH7777, or McA cells), an established model of mammalian lipoprotein metabolism, and mice. FITM2 expression was reduced using siRNA in cells and by liver specific cre-recombinase mediated deletion of the Fitm2 gene in mice. Effects of FITM2 deficiency on VLDL assembly and secretion in vitro and in vivo were measured by multiple methods, including density gradient ultracentrifugation, chromatography, mass spectrometry, stimulated Raman scattering (SRS) microscopy, sub-cellular fractionation, immunoprecipitation, immunofluorescence, and electron microscopy. MAIN FINDINGS 1) FITM2-deficient hepatic cells in vitro and in vivo secrete TG-depleted VLDL particles, but the number of particles is unchanged compared to controls; 2) FITM2 deficiency in mice on a high fat diet (HFD) results in decreased plasma TG levels. The number of apoB100-containing lipoproteins remains similar, but shift from VLDL to low density lipoprotein (LDL) density; 3) Both in vitro and in vivo, when TG synthesis is stimulated and FITM2 is deficient, TG accumulates in the ER, and despite its availability this pool is unable to fully lipidate apoB100 particles; 4) FITM2 deficiency disrupts ER morphology and results in ER stress. PRINCIPAL CONCLUSIONS The results suggest that FITM2 contributes to VLDL lipidation, especially when newly synthesized hepatic TG is in abundance. In addition to its fundamental importance in VLDL assembly, the results also suggest that under dysmetabolic conditions, FITM2 may be an important factor in the partitioning of TG between cytosolic LDs and VLDL particles.
Collapse
Affiliation(s)
- Haizhen Wang
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA; College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Cyrus Nikain
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA; Chemical Biology Program, Memorial Sloan Kettering Cancer Center and Weill Graduate School of Medical Sciences, Cornell University, NY, USA
| | - Konstantinos I Fortounas
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA
| | - Jaime Amengual
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA; Department of Food Sciences and Human Nutrition, University of Illinois, Urbana-Champaign, IL, USA
| | - Ozlem Tufanli
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA
| | - Maxwell La Forest
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA
| | - Yong Yu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA; State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Meng C Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA; Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Russell Watts
- Department of Pediatrics and Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Richard Lehner
- Department of Pediatrics and Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Yunping Qiu
- Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, NY, USA
| | - Min Cai
- Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, NY, USA
| | - Irwin J Kurland
- Stable Isotope and Metabolomics Core Facility, Albert Einstein College of Medicine, NY, USA
| | - Ira J Goldberg
- Department of Medicine (Endocrinology), NYU Grossman School of Medicine, NY, USA
| | - Sujith Rajan
- Department of Foundations of Medicine and Diabetes and Obesity Research Center, NYU Grossman Long Island School of Medicine, Mineola, NY, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine and Diabetes and Obesity Research Center, NYU Grossman Long Island School of Medicine, Mineola, NY, USA
| | - Jeffrey L Brodsky
- Department of Biological Sciences and the Center for Protein Conformational Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Edward A Fisher
- Department of Medicine (Cardiology), the Cardiovascular Research Center, and the Marc and Ruti Bell Program in Vascular Biology, NYU Grossman School of Medicine, NY, USA.
| |
Collapse
|
10
|
He L, Li H, Tang Y, Ren TB, Yuan L. Recent advances in fluorescent probes for fatty liver imaging by detecting lipid droplets. J Mater Chem B 2024; 12:10149-10162. [PMID: 39282742 DOI: 10.1039/d4tb01741b] [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: 10/18/2024]
Abstract
Fatty liver, a major health problem worldwide, is closely associated with aberrant accumulation and alteration of energy storage organelles, lipid droplets (LDs), in the disease process. Fluorescent probes with excellent optical performance, high sensitivity/selectivity and real-time monitoring have emerged as an attractive tool for the detection of LDs used in the diagnosis of fatty liver. In this review, we summarize various probes based on different response mechanisms to image LDs in the fatty liver process using different excitation imaging modes and emission wavelengths, including the visible to the near-infrared, two/three-photon, and the second near-infrared region. The perspectives and barriers associated with the reported lipid droplet (LD) probes for future development are also discussed.
Collapse
Affiliation(s)
- Long He
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 411201, P. R. China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China.
| | - Hang Li
- Key Laboratory of Theoretical Organic Chemistry and Functional Molecule, Ministry of Education, School of Chemistry and Chemical Engineering, Hunan University of Science and Technology, Xiangtan, 411201, P. R. China
| | - Yao Tang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China.
| | - Tian-Bing Ren
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China.
| | - Lin Yuan
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, P. R. China.
| |
Collapse
|
11
|
Zhao L, Jiang Q, Lei J, Cui J, Pan X, Yue Y, Zhang B. Bile acid disorders and intestinal barrier dysfunction are involved in the development of fatty liver in laying hens. Poult Sci 2024; 103:104422. [PMID: 39418789 DOI: 10.1016/j.psj.2024.104422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 10/19/2024] Open
Abstract
The pathogenesis of fatty liver is highly intricate. The role of the gut-liver axis in the development of fatty liver has gained increasing recognition in recent years. This study was conducted to explore the role of bile acid signaling and gut barrier in the pathogenesis of fatty liver. A total of 100 "Jing Tint 6" laying hens, 56-week-old, were used and fed basal diets until 60 weeks of age. At the end of the experiment, thirty individuals were selected based on the degree of hepatic steatosis. The hens with minimal hepatic steatosis (< 5 %) were chosen as healthy controls, while those with severe steatosis (> 33 %) in the liver were classified as the fatty liver group. Laying hens with fatty liver and healthy controls showed significant differences in body weight, liver index, abdominal fat ratio, feed conversion ratio (FCR), albumin height, Haugh unit, and biochemical indexes. The results of bile acid metabolomics revealed a clear separation in hepatic bile acid profiles between the fatty liver group and healthy controls, and multiple secondary bile acids were decreased in the fatty liver group, indicating disordered bile acid metabolism. Additionally, the mRNA levels of farnesoid X receptor (FXR) and genes related to bile acid transport were significantly decreased in both the liver and terminal ileum of hens with fatty liver. Moreover, the laying hens with fatty liver exhibited significant decreases in ileal crypt depth, the number of goblet cells, and the mRNA expression of tight junction-related proteins, alongside a significant increase in ileal permeability. Collectively, these findings suggest that disordered bile acids, suppressed FXR-mediated signaling, and impaired intestinal barrier function are potential factors promoting the development of fatty liver. These insights indicate that regulating bile acids and enhancing intestinal barrier function may become new preventive and therapeutic strategies for fatty liver in the near future.
Collapse
Affiliation(s)
- Lihua Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Qiuyu Jiang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiaqi Lei
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jian Cui
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xianjie Pan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuan Yue
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Bingkun Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
12
|
Hong P, Wang D, Wu Y, Zhang Q, Liu P, Pan J, Yu M, Tian W. A novel long noncoding RNA AK029592 contributes to thermogenic adipocyte differentiation. Stem Cells Transl Med 2024; 13:985-1000. [PMID: 39115701 PMCID: PMC11465168 DOI: 10.1093/stcltm/szae056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/29/2024] [Indexed: 10/11/2024] Open
Abstract
Exploration of factors originating from brown adipose tissue that govern the thermogenic adipocyte differentiation is imperative for comprehending the regulatory framework underlying brown fat biogenesis and for devising therapeutic approaches for metabolic disorders associated with obesity. Prior evidence has illuminated the pivotal role of long noncoding RNAs (lncRNAs) in orchestrating thermogenesis within adipose tissue. Here, we aimed to explore and identify the critical lncRNA that could promote thermogenic adipocyte differentiation and to provide a novel strategy to treat obesity-related metabolic diseases in the future. In this study, through amalgamation with our previous lncRNA microarray data from small extracellular vesicles derived from BAT (sEV-BAT), we have identified sEV-BAT-enriched lncRNA AK029592 as a critical constituent of the thermogenic program, which actively fostered beige adipocyte differentiation and enhanced the thermogenic capacities of adipose tissue. Moreover, lncRNA AK029592 could sponge miR-199a-5p in adipocytes to stimulate thermogenic gene expression. Consequently, we concluded lncRNA AK029592 as a crucial lncRNA component of the thermogenic program that regulated beige adipocyte differentiation and white adipose tissue browning, thereby providing a novel therapeutic target and strategy in combating obesity and related metabolic diseases.
Collapse
Affiliation(s)
- Pengyu Hong
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Dianri Wang
- Department of Head and Neck Surgery, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, People’s Republic of China
| | - Yue Wu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Qi Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Pan Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Jian Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Mei Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| | - Weidong Tian
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Engineering Research Center of Oral Translational Medicine, Ministry of Education & National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, People’s Republic of China
| |
Collapse
|
13
|
Guo H, Zions VS, Law BA, Hewitt KC. Potential of Raman-Reflectance Combination in Quantifying Liver Steatosis and Fat Droplet Size: Evidence From Monte Carlo Simulations and Phantom Studies. JOURNAL OF BIOPHOTONICS 2024; 17:e202400156. [PMID: 39223068 DOI: 10.1002/jbio.202400156] [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: 04/14/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
This study explores a combined strategy of Raman and reflectance spectroscopy for quantifying liver fat content and fat droplet size, crucial in assessing donor livers. By using Monte Carlo simulations and experimental setups with oil-in-water phantoms, our findings indicate that Raman scattering can solely differentiate between varying fat contents. At the same time, reflectance intensity is influenced by both fat content and oil droplet size, with a more pronounced sensitivity to fat droplet size. This study demonstrates the efficacy of combined Raman and reflectance spectroscopy in assessing liver steatosis and fat droplet size, potentially aiding in assessing donor livers for transplantation.
Collapse
Affiliation(s)
- Hao Guo
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medical Physics, Nova Scotia Health Authority, Halifax, Nova Scotia, Canada
| | - Vanessa S Zions
- Fisheries and Oceans Canada, Bedford Institute of Oceanography, Dartmouth, Nova Scotia, Canada
| | - Brent A Law
- Fisheries and Oceans Canada, Bedford Institute of Oceanography, Dartmouth, Nova Scotia, Canada
| | - Kevin C Hewitt
- Department of Physics and Atmospheric Science, Dalhousie University, Halifax, Nova Scotia, Canada
| |
Collapse
|
14
|
Corbo JH, Chung J. Mechanisms of lipid droplet degradation. Curr Opin Cell Biol 2024; 90:102402. [PMID: 39053179 DOI: 10.1016/j.ceb.2024.102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Lipid droplets (LDs) are subcellular organelles that play an integral role in lipid metabolism by regulating the storage and release of fatty acids, which are essential for energy production and various cellular processes. Lipolysis and lipophagy are the two major LD degradation pathways that mediate the utilization of lipids stored in these organelles. Recent studies have further uncovered alternative pathways, including direct lysosomal LD degradation and LD exocytosis. Here, we highlight recent findings that dissect the molecular basis of these diverse LD degradation pathways. Then, we discuss speculations on the crosstalk among these pathways and the potential unconventional roles of LD degradation.
Collapse
Affiliation(s)
- J H Corbo
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - J Chung
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
15
|
Yuan S, Zhao E. Recent advances of lipid droplet-targeted AIE-active materials for imaging, diagnosis and therapy. Biosens Bioelectron 2024; 267:116802. [PMID: 39332250 DOI: 10.1016/j.bios.2024.116802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 08/25/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Lipid droplets (LDs) are cellular organelles specialized in the storage and regulating the release of lipids critical for energy metabolism. As investigation on LDs deepens, the complex biological functions of LDs are revealed and their relationships with various diseases such as atherosclerosis, fatty liver, obesity, and cancer are uncovered. Fluorescence-based techniques with simple operations, visible results and high non-invasiveness are ideal tools for investigating LD-related biological processes and diseases. Materials with aggregation-induced emission (AIE) characteristics have emerged as promising candidates for investigating LDs due to their high signal-to-noise ratio (S/N), strong photostability, and large Stokes shift. This review discusses the principles and advantages of LD-targeting AIE probes for imaging LDs, diagnosis of LD-associated diseases including atherosclerotic plaques, liver diseases, acute kidney diseases and cancer, therapies with LD-targeting AIE-active photosensitizers and other relevant fields in the past five years. Through typical examples, we illustrate the status of investigating LD-related imaging, diagnosis of diseases and therapy with AIE materials. This review is expected to attract attentions from scientists with different research backgrounds and contribute to the further development of LD-targeting AIE materials.
Collapse
Affiliation(s)
- Sisi Yuan
- School of Science, Harbin Institute of Technology, Shenzhen, Guangdong, 518055, China
| | - Engui Zhao
- School of Science, Harbin Institute of Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
16
|
Hou H, Ji Y, Pan Y, Wang L, Liang Y. Persistent organic pollutants and metabolic diseases: From the perspective of lipid droplets. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 362:124980. [PMID: 39293651 DOI: 10.1016/j.envpol.2024.124980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 08/12/2024] [Accepted: 09/15/2024] [Indexed: 09/20/2024]
Abstract
The characteristic of semi-volatility enables persistent organic pollutants (POPs) almost ubiquitous in the environment. There is increasing concern about the potential risks of exposure to POPs due to their lipophilicity and readily bioaccumulation. Lipid droplets (LDs) are highly dynamic lipid storage organelles, alterations of intracellular LDs play a vital role in the progression of many prevalent metabolic diseases, such as type 2 diabetes (T2D) and nonalcoholic fatty liver disease (NAFLD). This article systematically reviewed the biological processes involved in LDs metabolism, the role of LDs proteins and LDs in metabolic diseases, and summarized updating researches on involvement of POPs in the progression of LDs-related metabolic diseases and potential mechanisms. POPs might change the physiological functions of LDs, also interfere the processes of adipogenesis and lipolysis by altering LDs synthesis, decomposition and function. However, further studies are still needed to explore the underlying mechanism of POPs-induced metabolic diseases, which can offer scientific evidences for metabolic disease prevention.
Collapse
Affiliation(s)
- Huixin Hou
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China
| | - Yaoting Ji
- Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yu Pan
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China
| | - Ling Wang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China.
| | - Yong Liang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, 430056, China
| |
Collapse
|
17
|
Rong J, Zhang Z, Peng X, Li P, Zhao T, Zhong Y. Mechanisms of hepatic and renal injury in lipid metabolism disorders in metabolic syndrome. Int J Biol Sci 2024; 20:4783-4798. [PMID: 39309427 PMCID: PMC11414397 DOI: 10.7150/ijbs.100394] [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: 07/02/2024] [Accepted: 08/13/2024] [Indexed: 09/25/2024] Open
Abstract
Metabolic syndrome (MetS) is a group of metabolic abnormalities that identifies people at risk for diabetes and cardiovascular disease. MetS is characterized by lipid disorders, and non-alcoholic fatty liver disease (NAFLD) and diabetic kidney disease (DKD) are thought to be the common hepatic and renal manifestations of MetS following abnormal lipid metabolism. This paper reviews the molecular mechanisms of lipid deposition in NAFLD and DKD, highlighting the commonalities and differences in lipid metabolic pathways in NAFLD and DKD. Hepatic and renal steatosis is the result of lipid acquisition exceeding lipid processing, i.e., fatty acid uptake and lipid regeneration exceed fatty acid oxidation and export. This process is directly regulated by the interactions of nuclear receptors, transporter proteins and transcription factors, whereas pathways such as oxidative stress, autophagy, cellular pyroptosis and gut flora are also key regulatory hubs for lipid metabolic homeostasis but act slightly differently in the liver and kidney. Such insights based on liver-kidney similarities and differences offer potential options for improved treatment.
Collapse
Affiliation(s)
- Jin Rong
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Beijing, PR China
- College of Life Science and Technology, Shandong Second Medical University, Weifang, Shandong, PR China
| | - Zixuan Zhang
- Department of Nephrology A, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Xiaoyu Peng
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Beijing, PR China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, PR China
| | - Ping Li
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Beijing, PR China
| | - Tingting Zhao
- Institute of Clinical Medical Sciences, State Key Laboratory of Respiratory Health and Multimorbidity, China-Japan Friendship Hospital, Beijing, PR China
| | - Yifei Zhong
- Department of Nephrology A, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| |
Collapse
|
18
|
Li K, Wang Y, Li X, Wang H. Comparative analysis of bile acid composition and metabolism in the liver of Bufo gargarizans aquatic larvae and terrestrial adults. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101322. [PMID: 39260083 DOI: 10.1016/j.cbd.2024.101322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 09/05/2024] [Accepted: 09/06/2024] [Indexed: 09/13/2024]
Abstract
Bile acids are crucial for lipid metabolism and their composition and metabolism differ among species. However, there have been no data on the differences in the composition and metabolism of bile acids between aquatic larvae and terrestrial adults of amphibians. This study explored the differences in composition and metabolism of bile acid between Bufo gargarizans larvae and adults. The results demonstrated that adult liver had a lower total bile acid level and a higher conjugated/total bile acid ratio than larval liver. Meanwhile, histological analysis revealed that the larvae showed a larger cross-sectional area of bile canaliculi lumen compared with the adults. The transcriptomic analysis showed that B. gargarizans larvae synthesized bile acids through both the alternative and the 24-hydroxylase pathway, while adults only synthesized bile acids through the 24-hydroxylase pathway. Moreover, bile acid regulator-related genes FXR and RXRα were highly expressed in adult, whereas genes involved in bile acid synthesis (CYP27A1 and CYP46A1) were highly expressed in larvae. The present study will provide valuable insights into understanding metabolic disorders and exploring novel bile acid-based therapeutics.
Collapse
Affiliation(s)
- Kaiyue Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Yufei Wang
- School of Biological Sciences, College of Science and Engineering, The University of Edinburgh, United Kingdom
| | - Xinyi Li
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
19
|
Anwar C, Lin JR, Tsai ML, Ho CT, Lai CS. Calebin A attenuated inflammation in RAW264.7 macrophages and adipose tissue to improve hepatic glucose metabolism and hyperglycemia in high-fat diet-fed obese mice. Eur J Pharmacol 2024; 978:176789. [PMID: 38945287 DOI: 10.1016/j.ejphar.2024.176789] [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: 04/03/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
The increased incidence of obesity, which become a global health problem, requires more functional food products with minor side and excellent effects. Calebin A (CbA) is a non-curcuminoid compound, which is reported to be an effective treatment for lipid metabolism and thermogenesis. However, its ability and mechanism of action in improving obesity-associated hyperglycemia remain unclear. This study was designed to explore the effect and mechanism of CbA in hyperglycemia via improvement of inflammation and glucose metabolism in the adipose tissue and liver in high-fat diet (HFD)-fed mice. After 10 weeks fed HFD, obese mice supplemented with CbA (25 and 100 mg/kg) for another 10 weeks showed a remarkable reducing adiposity and blood glucose. CbA modulated M1/M2 macrophage polarization, ameliorated inflammatory cytokines, and restored adiponectin as well as Glut 4 expression in the adipose tissue. In the in vitro study, CbA attenuated pro-inflammatory markers while upregulated anti-inflammatory IL-10 in LPS + IFNγ-generated M1 phenotype macrophages. In the liver, CbA attenuated steatosis, inflammatory infiltration, and protein levels of inflammatory TNF-α and IL-6. Moreover, CbA markedly upregulated Adiponectin receptor 1, AMPK, and insulin downstream Akt signaling to improve glycogen content and increase Glut2 protein. These findings indicated that CbA may be a novel therapeutic approach to treat obesity and hyperglycemia phenotype targeting on adipose inflammation and hepatic insulin signaling.
Collapse
Affiliation(s)
- Choirul Anwar
- Institute of Aquatic Science and Technology, Collage of Hydrosphere Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Jing-Ru Lin
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Mei-Ling Tsai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, 08901, USA.
| | - Ching-Shu Lai
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, 81157, Taiwan.
| |
Collapse
|
20
|
Wang B, Xi F, Jin C, Zhu HL, Tu M, Li Z. A ferrous fluorescence lifetime response probe for monitoring changes in lipid droplets during ferroptosis and imaging in liver disease model. Biosens Bioelectron 2024; 267:116742. [PMID: 39243450 DOI: 10.1016/j.bios.2024.116742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Ferrous ions (Fe2⁺) accumulation and abnormal alterations in lipid droplets (LDs) are closely associated with ferroptosis. In the liver, excessive iron accumulation promotes oxidative stress and exacerbates lipid droplet accumulation, while the disruption of iron homeostasis may also affect the formation and size of lipid droplets, their increased number and size can exacerbate the severity of disease under fatty liver conditions. The leads to hepatocyte damage, further triggering liver inflammation, fibrosis, and ultimately resulting in cirrhosis and hepatocellular carcinoma. Therefore, real-time monitoring of iron ion and lipid droplet changes is crucial for assessing the severity of liver disease, disease progression, and understanding the mechanisms of ferroptosis. We have developed a fluorescent probe, NRFep, for real-time monitoring of iron ion fluctuations and visualization of lipid droplet changes in ferroptosis and liver disease models. NRFep is specific and sensitive to iron ions and exhibits excellent stability in both cells and animal models. In addition, NRFep can be used to monitor changes in iron ions and lipid droplets in mouse liver injury and fatty liver models. Through fluorescence lifetime imaging technology, NRFep can also study the dynamic changes of intracellular iron ion content. NRFep provides a powerful tool for studying ferroptosis and related diseases, and its unique dual-monitoring function opens up new possibilities for developing new diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Bin Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Fangmin Xi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Chen Jin
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Hai-Liang Zhu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China
| | - Min Tu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Zhen Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, No.163 Xianlin Road, Nanjing, 210023, China.
| |
Collapse
|
21
|
Zhu J, Guo J, Liu Z, Liu J, Yuan A, Chen H, Qiu J, Dou X, Lu D, Le Y. Salvianolic acid A attenuates non-alcoholic fatty liver disease by regulating the AMPK-IGFBP1 pathway. Chem Biol Interact 2024; 400:111162. [PMID: 39047806 DOI: 10.1016/j.cbi.2024.111162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects approximately a quarter of the population and, to date, there is no approved drug therapy for this condition. Individuals with type 2 diabetes mellitus (T2DM) are at a significantly elevated risk of developing NAFLD, underscoring the urgency of identifying effective NAFLD treatments for T2DM patients. Salvianolic acid A (SAA) is a naturally occurring phenolic acid that is an important component of the water-soluble constituents isolated from the roots of Salvia miltiorrhiza Bunge. SAA has been demonstrated to possess anti-inflammatory and antioxidant stress properties. Nevertheless, its potential in ameliorating diabetes-associated NAFLD has not yet been fully elucidated. In this study, diabetic ApoE-/- mice were employed to establish a NAFLD model via a Western diet. Following this, they were treated with different doses of SAA (10 mg/kg, 20 mg/kg) via gavage. The study demonstrated a marked improvement in liver injury, lipid accumulation, inflammation, and the pro-fibrotic phenotype after the administration of SAA. Additionally, RNA-seq analysis indicated that the primary pathway by which SAA alleviates diabetes-induced NAFLD involves the cascade pathways of lipid metabolism. Furthermore, SAA was found to be effective in the inhibition of lipid accumulation, mitochondrial dysfunction and ferroptosis. A functional enrichment analysis of RNA-seq data revealed that SAA treatment modulates the AMPK pathway and IGFBP-1. Further experimental results demonstrated that SAA is capable of inhibiting lipid accumulation through the activation of the AMPK pathway and IGFBP-1.
Collapse
Affiliation(s)
- Ji Zhu
- The Third Affiliated Hospital of Zhejiang Chinese Medical University (Zhongshan Hospital of Zhejiang Province), Hangzhou, 330106, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jianan Guo
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Zhijun Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jing Liu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Aini Yuan
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Hang Chen
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jiannan Qiu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Xiaobing Dou
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Dezhao Lu
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yifei Le
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China; Lipid Metabolism Institute (Molecular Medicine Institute), Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
22
|
Buziau AM, Oosterveer MH, Wouters K, Bos T, Tolan DR, Agius L, Ford BE, Cassiman D, Stehouwer CDA, Schalkwijk CG, Brouwers MCGJ. Hepatic glucokinase regulatory protein and carbohydrate response element binding protein attenuation reduce de novo lipogenesis but do not mitigate intrahepatic triglyceride accumulation in Aldob deficiency. Mol Metab 2024; 87:101984. [PMID: 38972375 PMCID: PMC11300931 DOI: 10.1016/j.molmet.2024.101984] [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: 04/01/2024] [Revised: 06/15/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
OBJECTIVE Stable isotope studies have shown that hepatic de novo lipogenesis (DNL) plays an important role in the pathogenesis of intrahepatic lipid (IHL) deposition. Furthermore, previous research has demonstrated that fructose 1-phosphate (F1P) not only serves as a substrate for DNL, but also acts as a signalling metabolite that stimulates DNL from glucose. The aim of this study was to elucidate the mediators of F1P-stimulated DNL, with special focus on two key regulators of intrahepatic glucose metabolism, i.e., glucokinase regulatory protein (GKRP) and carbohydrate response element binding protein (ChREBP). METHODS Aldolase B deficient mice (Aldob-/-), characterized by hepatocellular F1P accumulation, enhanced DNL, and hepatic steatosis, were either crossed with GKRP deficient mice (Gckr-/-) or treated with short hairpin RNAs directed against hepatic ChREBP. RESULTS Aldob-/- mice showed higher rates of de novo palmitate synthesis from glucose when compared to wildtype mice (p < 0.001). Gckr knockout reduced de novo palmitate synthesis in Aldob-/- mice (p = 0.017), without affecting the hepatic mRNA expression of enzymes involved in DNL. In contrast, hepatic ChREBP knockdown normalized the hepatic mRNA expression levels of enzymes involved in DNL and reduced fractional DNL in Aldob-/- mice (p < 0.05). Of interest, despite downregulation of DNL in response to Gckr and ChREBP attenuation, no reduction in intrahepatic triglyceride levels was observed. CONCLUSIONS Both GKRP and ChREBP mediate F1P-stimulated DNL in aldolase B deficient mice. Further studies are needed to unravel the role of GKRP and hepatic ChREBP in regulating IHL accumulation in aldolase B deficiency.
Collapse
Affiliation(s)
- Amée M Buziau
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Maaike H Oosterveer
- Department of Pediatrics, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Kristiaan Wouters
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Trijnie Bos
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Dean R Tolan
- Department of Biology, Boston University, Boston, MA, USA
| | - Loranne Agius
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Brian E Ford
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - David Cassiman
- Department of Gastroenterology-Hepatology and Metabolic Center, University Hospital Leuven, Leuven, Belgium
| | - Coen D A Stehouwer
- Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Casper G Schalkwijk
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands
| | - Martijn C G J Brouwers
- Department of Internal Medicine, Division of Endocrinology and Metabolic Disease, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, the Netherlands; Department of Internal Medicine, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
23
|
Wu J, Zhang Y, Wu X, Chen T, Yan M, Shi S, Zhang F, Fan B, Zhao B, Cheng H. Near infrared aggregation-induced emission fluorescent materials for lipid droplets testing and photodynamic therapy. LUMINESCENCE 2024; 39:e4885. [PMID: 39238366 DOI: 10.1002/bio.4885] [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: 07/01/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
Near-infrared (NIR) fluorescent probes with aggregation-induced emission (AIE) properties are of great significance in cell imaging and cancer therapy. However, the complexity of its synthesis, poor photostabilities, and expensive raw materials still pose some obstacles to their practical application. This study reported an AIE luminescent material with red emission and its application in in vitro imaging and photodynamic therapy (PDT) study. This material has the characteristics of simple synthesis, large Stokes shift, good photostabilities, and excellent lipid droplets-specific testing ability. Interestingly, this red-emitting material can effectively produce reactive oxygen species (ROS) under white light irradiation, further achieving PDT-mediated killing of cancer cells. In conclusion, this study demonstrates a simple approach to synthesize NIR AIE probes with both imaging and therapeutic effects, providing an ideal architecture for constructing long-wavelength emission AIE materials.
Collapse
Affiliation(s)
- Jiang Wu
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, China
- Xianning Public Inspection and Testing Center, Xianning, China
| | - Yao Zhang
- School of Health Service and Management, Shanxi University of Chinese Medicine, Taiyuan, China
| | - Xiaoxiao Wu
- Xianning Public Inspection and Testing Center, Xianning, China
| | - Tu Chen
- Xianning Public Inspection and Testing Center, Xianning, China
| | - Miao Yan
- Department of Chemistry, Xinzhou Normal University, Xinzhou, China
| | - Shijing Shi
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, China
| | - Fei Zhang
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, China
| | - Baolei Fan
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, China
| | - Baoqing Zhao
- Medicine Research Institute & Hubei Key Laboratory of Diabetes and Angiopathye, Hubei University of Science and Technology, Xianning, China
| | - Hong Cheng
- Hubei Key Laboratory of Radiation Chemistry and Functional Materials, School of Nuclear Technology and Chemistry & Biology, Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
24
|
Zheng Z, Ling X, Li Y, Qiao S, Zhang S, Wu J, Ma Z, Li M, Guo X, Li Z, Feng Y, Liu X, Goodfellow IG, Zheng H, Xiao S. Host cells reprogram lipid droplet synthesis through YY1 to resist PRRSV infection. mBio 2024; 15:e0154924. [PMID: 38953350 PMCID: PMC11323570 DOI: 10.1128/mbio.01549-24] [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: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 07/04/2024] Open
Abstract
Metabolism in host cells can be modulated after viral infection, favoring viral survival or clearance. Here, we report that lipid droplet (LD) synthesis in host cells can be modulated by yin yang 1 (YY1) after porcine reproductive and respiratory syndrome virus (PRRSV) infection, resulting in active antiviral activity. As a ubiquitously distributed transcription factor, there was increased expression of YY1 upon PRRSV infection both in vitro and in vivo. YY1 silencing promoted the replication of PRRSV, whereas YY1 overexpression inhibited PRRSV replication. PRRSV infection led to a marked increase in LDs, while YY1 knockout inhibited LD synthesis, and YY1 overexpression enhanced LD accumulation, indicating that YY1 reprograms PRRSV infection-induced intracellular LD synthesis. We also showed that the viral components do not colocalize with LDs during PRRSV infection, and the effect of exogenously induced LD synthesis on PRRSV replication is nearly lethal. Moreover, we demonstrated that YY1 affects the synthesis of LDs by regulating the expression of lipid metabolism genes. YY1 negatively regulates the expression of fatty acid synthase (FASN) to weaken the fatty acid synthesis pathway and positively regulates the expression of peroxisome proliferator-activated receptor gamma (PPARγ) to promote the synthesis of LDs, thus inhibiting PRRSV replication. These novel findings indicate that YY1 plays a crucial role in regulating PRRSV replication by reprogramming LD synthesis. Therefore, our study provides a novel mechanism of host resistance to PRRSV and suggests potential new antiviral strategies against PRRSV infection.IMPORTANCEPorcine reproductive and respiratory virus (PRRSV) has caused incalculable economic damage to the global pig industry since it was first discovered in the 1980s. However, conventional vaccines do not provide satisfactory protection. It is well known that viruses are parasitic pathogens, and the completion of their replication life cycle is highly dependent on host cells. A better understanding of host resistance to PRRSV infection is essential for developing safe and effective strategies to control PRRSV. Here, we report a crucial host antiviral molecule, yin yang 1 (YY1), which is induced to be expressed upon PRRSV infection and subsequently inhibits virus replication by reprogramming lipid droplet (LD) synthesis through transcriptional regulation. Our work provides a novel antiviral mechanism against PRRSV infection and suggests that targeting YY1 could be a new strategy for controlling PRRSV.
Collapse
Affiliation(s)
- Zifang Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xue Ling
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yang Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shuang Qiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Shuangquan Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jie Wu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Zhiqian Ma
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Mingyu Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xuyang Guo
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhiwei Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Yingtong Feng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ian G. Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - Haixue Zheng
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Shuqi Xiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| |
Collapse
|
25
|
Jing Y, Cao RX, Lei X, Wang ZL, Huang XL, Di JR, Mi ZX, Zhao X, Wang M, Jiang MM, Yang WZ, Li X, Miao L, Zhang H, Zhang P. Structural characterization of polysaccharide from the peel of Trichosanthes kirilowii Maxim and its anti-hyperlipidemia activity by regulating gut microbiota and inhibiting cholesterol absorption. Bioorg Chem 2024; 149:107487. [PMID: 38805910 DOI: 10.1016/j.bioorg.2024.107487] [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: 04/25/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 05/30/2024]
Abstract
The peel of Trichosanthes kirilowii Maxim, is considered one of the primary sources for Trichosanthis pericarpium in traditional Chinese medicine, exhibiting lipid-lowering properties. The impact on hyperlipidemia mice of the crude polysaccharide from the peel of T. Kirilowii (TRP) was investigated in this study. The findings revealed that TRP exhibited a significant improvement in hepatic lipid deposition. Moreover, it significantly decreased serum levels of TC, TG, and LDL-C, while concurrently increasing HDL-C. 16S rRNA amplicon sequencing technique revealed that TRP group exhibited an increased relative abundance of Actinobacteria, a down-regulated relative abundance of Ruminiclostridium, and an up-regulated relative abundance of Ileibacterium. Therefore, TRP might play a role in anti-hyperlipidemia through regulation of the intestinal milieu and enhancement of microbial equilibrium. Consequently, targeted fractionation of TRP resulted in the isolation of a homogeneous acidic polysaccharide termed TRP-1. The TRP-1 polysaccharide, with an average molecular weight of 1.00 × 104 Da, and was primarily composed of Rha, GlcA, GalA, Glc, Gal and Ara. TRP-1 possessed a backbone consisting of alternating connections between → 6)-α-Galp-(1 → 4)-α-Rhap-(1 → 6)-α-Galp-(2 → 6)-β-Galp-(1 → 6)-α-Galp-(2 → 6)-β-Galp-(1 → units and branched chain containing → 6)-α-Glcp-(1→, 2,4)-β-Glcp-(1, and → 4)-α-GlapA-(1→. Both TRP and TRP-1 exhibited significant disruption of cholesterol micelles, highlighting their potential as lipid-lowering agents that effectively inhibit cholesterol absorption pathways.
Collapse
Affiliation(s)
- Yi Jing
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruo-Xin Cao
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xi Lei
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ze-Ling Wang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiang-Long Huang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jing-Rui Di
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zhuo-Xin Mi
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xin Zhao
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Meng Wang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Miao-Miao Jiang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wen-Zhi Yang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xue Li
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Lin Miao
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Han Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Peng Zhang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Ministry of Education Key Laboratory of Pharmacology of Traditional Chinese Medicine Formulae, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
26
|
Janmeda P, Jain D, Chaudhary P, Meena M, Singh D. A systematic review on multipotent carcinogenic agent, N-nitrosodiethylamine (NDEA), its major risk assessment, and precautions. J Appl Toxicol 2024; 44:1108-1128. [PMID: 38212177 DOI: 10.1002/jat.4574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 12/01/2023] [Accepted: 12/09/2023] [Indexed: 01/13/2024]
Abstract
The International Agency for Research on Cancer has classified N-nitrosodiethylamine (NDEA) as a possible carcinogen and mutagenic substances, placing it in category 2A of compounds that are probably harmful to humans. It is found in nature and tobacco smoke, along with its precursors, and is also synthesized endogenously in the human body. The oral or parenteral administration of a minimal quantity of NDEA results in severe liver and kidney organ damage. The NDEA required bioactivation by CYP450 enzyme to form DNA adduct in the alkylation mechanism. Thus, this bioactivation directs oxidative stress and injury to cells due to the higher formation of reactive oxygen species and alters antioxidant system in tissues, whereas free radical scavengers guard the membranes from NDEA-directed injury in many enzymes. This might be one of the reasons in the etiology of cancer that is not limited to a certain target organ but can affect various organs and organ systems. Although there are various possible approaches for the treatment of NDEA-induced cancer, their therapeutic outcomes are still very dismal. However, several precautions were considered to be taken during handling or working with NDEA, as it considered being the best way to lower down the occurrence of NDEA-directed cancers. The present review was designed to enlighten the general guidelines for working with NDEA, possible mechanism, to alter the antioxidant line to cause malignancy in different parts of animal body along with its protective agents. Thus, revelation to constant, unpredictable stress situations even in common life may remarkably augment the toxic potential through the rise in the oxidative stress and damage of DNA.
Collapse
Affiliation(s)
- Pracheta Janmeda
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk, Rajasthan, India
| | - Divya Jain
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk, Rajasthan, India
| | - Priya Chaudhary
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Tonk, Rajasthan, India
| | - Mukesh Meena
- Laboratory of Phytopathology and Microbial Biotechnology, Department of Botany, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| | - Devendra Singh
- Department of Chemistry, Mohanlal Sukhadia University, Udaipur, Rajasthan, India
| |
Collapse
|
27
|
Sun X, Yu Q, Qi Y, Kang B, Zhao X, Liu L, Wang P, Cong M, Liu T. Peridroplet mitochondria are associated with the severity of MASLD and the prevention of MASLD by diethyldithiocarbamate. J Lipid Res 2024; 65:100590. [PMID: 38981572 PMCID: PMC11381863 DOI: 10.1016/j.jlr.2024.100590] [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: 11/25/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/11/2024] Open
Abstract
Mitochondria can contact lipid droplets (LDs) to form peridroplet mitochondria (PDM) which trap fatty acids in LDs by providing ATP for triglyceride synthesis and prevent lipotoxicity. However, the role of PDM in metabolic dysfunction associated steatotic liver disease (MASLD) is not clear. Here, the features of PDM in dietary MASLD models with different severity in mice were explored. Electron microscope photographs show that LDs and mitochondria rarely come into contact with each other in normal liver. In mice fed with high-fat diet, PDM can be observed in the liver as early as the beginning of steatosis in hepatocytes. For the first time, we show that PDM in mouse liver varies with the severity of MASLD. PDM and cytosolic mitochondria were isolated from the liver tissue of MASLD and analyzed by quantitative proteomics. Compared with cytosolic mitochondria, PDM have enhanced mitochondrial respiration and ATP synthesis. Diethyldithiocarbamate (DDC) alleviates choline-deficient, L-amino acid-defined diet-induced MASLD, while increases PDM in the liver. Similarly, DDC promotes the contact of mitochondria-LDs in steatotic C3A cells in vitro. Meanwhile, DDC promotes triglyceride synthesis and improves mitochondrial dysfunction in MASLD. In addition, DDC upregulates perilipin 5 both in vivo and in vitro, which is considered as a key regulator in PDM formation. Knockout of perilipin 5 inhibits the contact of mitochondria-LDs induced by DDC in C3A cells. These results demonstrate that PDM might be associated with the progression of MASLD and the prevention of MASLD by DDC.
Collapse
Affiliation(s)
- Xiangyun Sun
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China
| | - Qinghong Yu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China
| | - Yifei Qi
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China
| | - Bilian Kang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China; Department of Hepatology, National Clinical Research Center for Digestive Diseases, Beijing, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China
| | - Lin Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China; Department of Hepatology, National Clinical Research Center for Digestive Diseases, Beijing, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China
| | - Ping Wang
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China; Department of Hepatology, National Clinical Research Center for Digestive Diseases, Beijing, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China; Department of Hepatology, National Clinical Research Center for Digestive Diseases, Beijing, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China
| | - Tianhui Liu
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China; Department of Hepatology, State Key Lab of Digestive Health, Beijing, China; Department of Hepatology, National Clinical Research Center for Digestive Diseases, Beijing, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, China.
| |
Collapse
|
28
|
Zhuang W, Li J, Qu T, Shao R, Chen J, Li S, Chen M, Wang Y. A lipid activated color switchable probe for the imaging of diseased aortic valves. Talanta 2024; 275:126069. [PMID: 38692042 DOI: 10.1016/j.talanta.2024.126069] [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: 02/21/2024] [Revised: 03/26/2024] [Accepted: 04/06/2024] [Indexed: 05/03/2024]
Abstract
Lipid deposition has been considered one of the key factors in the occurrence of valvular heart disease (VHD) and a great potential target for the diagnosis of VHD. However, the development of lipid imaging technologies and efficient lipid specific probes is in urgent demand. In this work, we have prepared a lipid droplets (LDs) targeted fluorescence probe CPTM based on a push-pull electronic structure for the imaging of diseased aortic valves. CPTM showed obvious twisted intramolecular charge transfer (TICT) effect and its emission changed from 600 nm in water to 508 nm in oil. CPTM not only exhibited good biocompatibility and high photostability, but also impressive LDs specific imaging performance in human primary valvular interstitial cells and human diseased aortic valves. Moreover, the dynamic changes of intracellular LDs could be monitor in real-time after staining with CPTM. These results were expected to offer new ideals for the designing of novel LDs specific probes for further bioimaging applications.
Collapse
Affiliation(s)
- Weihua Zhuang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China; National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610065, PR China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China
| | - Tianyi Qu
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China
| | - Ruochen Shao
- Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China
| | - Jingruo Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China; Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China
| | - Shufen Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China.
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China; Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Alley, Chengdu, 610041, PR China.
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, 29 Wangjiang Road, Chengdu, 610065, PR China.
| |
Collapse
|
29
|
Walls KM, Joh JY, Hong KU, Hein DW. Heterocyclic Amines Disrupt Lipid Homeostasis in Cryopreserved Human Hepatocytes. Cardiovasc Toxicol 2024; 24:747-756. [PMID: 38851663 PMCID: PMC11300155 DOI: 10.1007/s12012-024-09874-1] [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: 02/23/2024] [Accepted: 05/23/2024] [Indexed: 06/10/2024]
Abstract
Metabolic dysfunction associated-steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH) is the liver manifestation of metabolic syndrome, which is characterized by insulin resistance, hyperglycemia, hypertension, dyslipidemia, and/or obesity. Environmental pollutant exposure has been recently identified as a risk factor for developing MASH. Heterocyclic amines (HCAs) are mutagens generated when cooking meat at high temperatures or until well-done. Recent epidemiological studies reported that dietary HCA exposure may be linked to insulin resistance and type II diabetes, and we recently reported that HCAs induce insulin resistance and glucose production in human hepatocytes. However, no previous studies have examined the effects of HCAs on hepatic lipid homeostasis. In the present study, we assessed the effects of two common HCAs, MeIQx (2-amino-3, 8-dimethylimidazo [4, 5-f] quinoxaline) and PhIP (2-amino-1-methyl-6-phenylimidazo[4, 5-b] pyridine), on lipid homeostasis in cryopreserved human hepatocytes. Exposure to a single concentration of 25 μM MeIQx or PhIP in human hepatocytes led to dysregulation of lipid homeostasis, typified by significant increases in lipid droplets and triglycerides. PhIP significantly increased expression of lipid droplet-associated genes, PNPLA3 and HSD17B13, and both HCAs significantly increased PLIN2. Exposure to MeIQx or PhIP also significantly increased expression of several key genes involved in lipid synthesis, transport and metabolism, including FASN, DGAT2, CPT1A, SCD, and CD36. Furthermore, both MeIQx and PhIP significantly increased intracellular cholesterol and decreased expression of PON1 which is involved in cholesterol efflux. Taken together, these results suggest that HCAs dysregulate lipid production, metabolism, and storage. The current study demonstrates, for the first time, that HCA exposure may lead to fat accumulation in hepatocytes, which may contribute to hepatic insulin resistance and MASH.
Collapse
Affiliation(s)
- Kennedy M Walls
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Environmental Justice, Community Health and Environmental Review Division, US Environmental Protection Agency, Chicago, USA
| | - Jonathan Y Joh
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
| | - Kyung U Hong
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA
- Department of Pharmaceutical and Administrative Sciences, Western New England University, Springfield, USA
| | - David W Hein
- Department of Pharmacology and Toxicology and Brown Cancer Center, University of Louisville School of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
30
|
Tzirkel-Hancock N, Raz C, Sharabi L, Argov-Argaman N. The Stressogenic Impact of Bacterial Secretomes Is Modulated by the Size of the Milk Fat Globule Used as a Substrate. Foods 2024; 13:2429. [PMID: 39123620 PMCID: PMC11312077 DOI: 10.3390/foods13152429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/28/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Milk fat globules (MFGs) are produced by mammary epithelial cells (MECs) and originate from intracellular lipid droplets with a wide size distribution. In the mammary gland and milk, bacteria can thrive on MFGs. Herein, we aimed to investigate whether the response of MECs to the bacterial secretome is dependent on the MFG size used as a substrate for the bacteria, and whether the response differs between pathogenic and commensal bacteria. We used secretomes from both Bacillus subtilis and E. coli. Proinflammatory gene expression in MECs was elevated by the bacteria secretomes from both bacteria sources, while higher expression was found in cells exposed to the secretome of bacteria grown on large MFGs. The secretome of B. subtilis reduced lipid droplet size in MECs. When the secretome originated from E. coli, lipid droplet size in MEC cytoplasm was elevated with a stronger response to the secretome from bacteria grown on large compared with small MFGs. These results indicate that MEC response to bacterial output is modulated by bacteria type and the size of MFGs used by the bacteria, which can modulate the stress response of the milk-producing cells, their lipid output, and consequently milk quality.
Collapse
Affiliation(s)
| | | | | | - Nurit Argov-Argaman
- Department of Animal Science, The Robert H Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot 7610001, Israel; (N.T.-H.)
| |
Collapse
|
31
|
Kang SY, Kimura M, Shrestha S, Lewis P, Lee S, Cai Y, Joshi P, Acharya P, Liu J, Yang Y, Sanchez JG, Ayyagari S, Alsberg E, Wells JM, Takebe T, Lee MY. A Pillar and Perfusion Plate Platform for Robust Human Organoid Culture and Analysis. Adv Healthc Mater 2024; 13:e2302502. [PMID: 37616035 PMCID: PMC10891301 DOI: 10.1002/adhm.202302502] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/10/2023] [Indexed: 08/25/2023]
Abstract
Human organoids have the potential to revolutionize in vitro disease modeling by providing multicellular architecture and function that are similar to those in vivo. This innovative and evolving technology, however, still suffers from assay throughput and reproducibility to enable high-throughput screening (HTS) of compounds due to cumbersome organoid differentiation processes and difficulty in scale-up and quality control. Using organoids for HTS is further challenged by the lack of easy-to-use fluidic systems that are compatible with relatively large organoids. Here, these challenges are overcome by engineering "microarray three-dimensional (3D) bioprinting" technology and associated pillar and perfusion plates for human organoid culture and analysis. High-precision, high-throughput stem cell printing, and encapsulation techniques are demonstrated on a pillar plate, which is coupled with a complementary deep well plate and a perfusion well plate for static and dynamic organoid culture. Bioprinted cells and spheroids in hydrogels are differentiated into liver and intestine organoids for in situ functional assays. The pillar/perfusion plates are compatible with standard 384-well plates and HTS equipment, and thus may be easily adopted in current drug discovery efforts.
Collapse
Affiliation(s)
- Soo-Yeon Kang
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - Masaki Kimura
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - Phillip Lewis
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sangjoon Lee
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - Yuqi Cai
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Pranav Joshi
- Bioprinting Laboratories Inc., Dallas, TX, 75234, USA
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - Jiafeng Liu
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - J Guillermo Sanchez
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Sriramya Ayyagari
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
- Departments of Orthopedics, Pharmacology, and Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - James M Wells
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Takanori Takebe
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
- Center for Stem Cell and Organoid Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
- Bioprinting Laboratories Inc., Dallas, TX, 75234, USA
| |
Collapse
|
32
|
Zhang Z, Yu Z, Liang D, Song K, Kong X, He M, Liao X, Huang Z, Kang A, Bai R, Ren Y. Roles of lipid droplets and related proteins in metabolic diseases. Lipids Health Dis 2024; 23:218. [PMID: 39030618 PMCID: PMC11264848 DOI: 10.1186/s12944-024-02212-y] [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: 04/12/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Lipid droplets (LDs), which are active organelles, derive from the monolayer membrane of the endoplasmic reticulum and encapsulate neutral lipids internally. LD-associated proteins like RAB, those in the PLIN family, and those in the CIDE family participate in LD formation and development, and they are active players in various diseases, organelles, and metabolic processes (i.e., obesity, non-alcoholic fatty liver disease, and autophagy). Our synthesis on existing research includes insights from the formation of LDs to their mechanisms of action, to provide an overview needed for advancing research into metabolic diseases and lipid metabolism.
Collapse
Affiliation(s)
- Zhongyang Zhang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Zhenghang Yu
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Dianyuan Liang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Ke Song
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Xiangxin Kong
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Ming He
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
| | - Xinxin Liao
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Ziyan Huang
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Aijia Kang
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Rubing Bai
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China.
| | - Yixing Ren
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China.
- General Surgery, Chengdu XinHua Hospital Affiliated to North Sichuan Medical College, Chengdu, 610000, China.
| |
Collapse
|
33
|
Léger T, Alilat S, Ferron PJ, Dec L, Bouceba T, Lanceleur R, Huet S, Devriendt-Renault Y, Parinet J, Clément B, Fessard V, Le Hégarat L. Chlordecone-induced hepatotoxicity and fibrosis are mediated by the proteasomal degradation of septins. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:135177. [PMID: 39018595 DOI: 10.1016/j.jhazmat.2024.135177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/08/2024] [Accepted: 07/09/2024] [Indexed: 07/19/2024]
Abstract
Chlordecone (CLD) is a pesticide persisting in soils and contaminating food webs. CLD is sequestered in the liver and poorly metabolized into chlordecol (CLDOH). In vitro liver cell models were used to investigate the fate and mechanistic effects of CLD and CLDOH using multiomics. A 3D-cell model was used to investigate whether CLD and CLDOH can affect susceptibility to the metabolic dysfunction-associated steatotic liver disease (MASLD). Hepatocytes were more sensitive to CLD than CLDOH. CLDOH was intensively metabolized into a glucuronide conjugate, whereas CLD was sequestered. CLD but not CLDOH induced a depletion of Septin-2,- 7,- 9,- 10,- 11 due to proteasomal degradation. Septin binding with CLD and CLDOH was confirmed by surface plasmon resonance. CLD disrupted lipid droplet size and increased saturated long-chain dicarboxylic acid production by inhibiting stearoyl-CoA desaturase (SCD) abundance. Neither CLD nor CLDOH induced steatosis, but CLD induced fibrosis in the 3D model of MASLD. To conclude, CLD hepatoxicity is specifically driven by the degradation of septins. CLDOH, was too rapidly metabolized to induce septin degradation. We show that the conversion of CLD to CLDOH reduced hepatotoxicity and fibrosis in liver organoids. This suggests that protective strategies could be explored to reduce the hepatotoxicity of CLD.
Collapse
Affiliation(s)
- Thibaut Léger
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France.
| | - Sarah Alilat
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Pierre-Jean Ferron
- INSERM, University of Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer) UMR_A 1317, UMR_S 1241, Previtox Network, 35000 Rennes, France
| | - Léonie Dec
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Tahar Bouceba
- Sorbonne University, CNRS, Institut de Biologie Paris-Seine (IBPS), Protein Engineering Platform, Molecular Interaction Service, Paris, France
| | - Rachelle Lanceleur
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Sylvie Huet
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Yoann Devriendt-Renault
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Pesticides and Marine Biotoxins (PBM) unit, Maison-Alfort Laboratory, 94701 Maison-Alfort CEDEX, France
| | - Julien Parinet
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Pesticides and Marine Biotoxins (PBM) unit, Maison-Alfort Laboratory, 94701 Maison-Alfort CEDEX, France
| | - Bruno Clément
- INSERM, University of Rennes, INRAE, Institut NuMeCan (Nutrition, Metabolisms and Cancer) UMR_A 1317, UMR_S 1241, Previtox Network, 35000 Rennes, France
| | - Valérie Fessard
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| | - Ludovic Le Hégarat
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Toxicology of Contaminants Unit, Fougères Laboratory, 35306 Fougères CEDEX, France
| |
Collapse
|
34
|
Nakamura A, Yoshimura T, Ichikawa T, Okuyama K. Prognostic significance of low hepatic fat content in advanced chronic liver disease: MRI-PDFF insights. Ann Hepatol 2024; 29:101507. [PMID: 38723748 DOI: 10.1016/j.aohep.2024.101507] [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: 11/08/2023] [Revised: 03/19/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024]
Abstract
INTRODUCTION AND OBJECTIVES The mechanisms of hepatic fat loss in late-stage metabolic dysfunction-associated fatty liver disease (MASLD) are enigmatic and the prognostic significance of low hepatic fat content (LHF) in chronic liver disease (CLD) is unknown. Proton density fat fraction (PDFF), measured by magnetic resonance imaging (MRI), is considered the most accurate noninvasive method for quantifying hepatic fat content. This study aimed to address these issues by evaluating PDFF. PATIENTS AND METHODS This is a single-center, retrospective study involving 762 patients with CLD, measuring liver stiffness (LS) using MR elastography and PDFF using MRI. LHF was defined as a PDFF ≤ 2.7 % and hepatic reserve function was assessed using the albumin-bilirubin (ALBI) score. Multivariate analysis explored associations between variables. RESULTS LHF was 27 % in the entire cohort, and PDFF was significantly decreased with LS ≥ 5.5 kPa (p < 0.05). On the multivariate analysis, low body mass index and ALBI score were independently associated with LHF (p < 0.05). In advanced CLD (n = 288), ALBI score and PDFF showed a significant negative correlation regardless of etiology (MASLD/non-MASLD: r= -0.613/-0.233), and the prevalence of LHF increased with progression of ALBI grade (p < 0.01 each). In addition, lower PDFF was associated with increased liver-related and all-cause mortality (p < 0.01), and Cox proportional hazards models extracted LHF as an independent prognostic factor, along with ALBI score and hepatocellular carcinoma (p < 0.05 each). CONCLUSIONS In ACLD, hepatic reserve dysfunction contributed to hepatic fat loss independent of nutritional status, suggesting that LHF may be a poor prognostic factor in all etiologies.
Collapse
Affiliation(s)
- Atsushi Nakamura
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan.
| | - Tsubasa Yoshimura
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Takeshi Ichikawa
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Keiji Okuyama
- Gastroenterological Liver Disease Center, Nippon Koukan Hospital, Kawasaki, Kanagawa, Japan
| |
Collapse
|
35
|
Risi R, Vidal-Puig A, Bidault G. An adipocentric perspective of pancreatic lipotoxicity in diabetes pathogenesis. J Endocrinol 2024; 262:e230313. [PMID: 38642584 PMCID: PMC11227041 DOI: 10.1530/joe-23-0313] [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: 09/30/2023] [Accepted: 04/16/2024] [Indexed: 04/22/2024]
Abstract
Obesity and diabetes represent two increasing and invalidating public health issues that often coexist. It is acknowledged that fat mass excess predisposes to insulin resistance and type 2 diabetes mellitus (T2D), with the increasing incidence of the two diseases significantly associated. Moreover, emerging evidence suggests that obesity might also accelerate the appearance of type 1 diabetes (T1D), which is now a relatively frequent comorbidity in patients with obesity. It is a common clinical finding that not all patients with obesity will develop diabetes at the same level of adiposity, with gender, genetic, and ethnic factors playing an important role in defining the timing of diabetes appearance. The adipose tissue (AT) expandability hypothesis explains this paradigm, indicating that the individual capacity to appropriately store energy surplus in the form of fat within the AT determines and prevents the toxic deposition of lipids in other organs, such as the pancreas. Thus, we posit that when the maximal storing capacity of AT is exceeded, individuals will develop T2D. In this review, we provide insight into mechanisms by which the AT controls pancreas lipid content and homeostasis in case of obesity to offer an adipocentric perspective of pancreatic lipotoxicity in the pathogenesis of diabetes. Moreover, we suggest that improving AT function is a valid therapeutic approach to fighting obesity-associated complications including diabetes.
Collapse
Affiliation(s)
- Renata Risi
- Department of Experimental Medicine, Sapienza University of Rome, Sapienza University of Rome, Rome, Italy
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P. R. China
- Centro de Investigacion Principe Felipe, Valencia, Spain
| | - Guillaume Bidault
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Cambridge, UK
| |
Collapse
|
36
|
Madsen S, Delgado AC, Cadilhac C, Maillard V, Battiston F, Igelbüscher CM, De Neck S, Magrinelli E, Jabaudon D, Telley L, Doetsch F, Knobloch M. A fluorescent perilipin 2 knock-in mouse model reveals a high abundance of lipid droplets in the developing and adult brain. Nat Commun 2024; 15:5489. [PMID: 38942786 PMCID: PMC11213871 DOI: 10.1038/s41467-024-49449-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: 07/20/2022] [Accepted: 06/03/2024] [Indexed: 06/30/2024] Open
Abstract
Lipid droplets (LDs) are dynamic lipid storage organelles. They are tightly linked to metabolism and can exert protective functions, making them important players in health and disease. Most LD studies in vivo rely on staining methods, providing only a snapshot. We therefore developed a LD-reporter mouse by labelling the endogenous LD coat protein perilipin 2 (PLIN2) with tdTomato, enabling staining-free fluorescent LD visualisation in living and fixed tissues and cells. Here we validate this model under standard and high-fat diet conditions and demonstrate that LDs are highly abundant in various cell types in the healthy brain, including neurons, astrocytes, ependymal cells, neural stem/progenitor cells and microglia. Furthermore, we also show that LDs are abundant during brain development and can be visualized using live imaging of embryonic slices. Taken together, our tdTom-Plin2 mouse serves as a novel tool to study LDs and their dynamics under both physiological and diseased conditions in all tissues expressing Plin2.
Collapse
Affiliation(s)
- Sofia Madsen
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | | | - Christelle Cadilhac
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Vanille Maillard
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Fabrice Battiston
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | | | - Simon De Neck
- Institute of Veterinary Pathology, University of Zurich, Zurich, Switzerland
| | - Elia Magrinelli
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Denis Jabaudon
- Department of Basic Neurosciences, University of Geneva, Geneva, Switzerland
| | - Ludovic Telley
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | | - Marlen Knobloch
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
37
|
Zhu Y, Yuan Y, Si H, Li S, Zhao F, Mu R, Lin Z, Wang X, Qiu Q, Xu C, Ji L, Li Z. Lipidomic and transcriptomic profiles provide new insights into the triacylglycerol and glucose handling capacities of the Arctic fox. Front Vet Sci 2024; 11:1388532. [PMID: 38988981 PMCID: PMC11233799 DOI: 10.3389/fvets.2024.1388532] [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: 04/04/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
The Arctic fox (Vulpes lagopus) is a species indigenous to the Arctic and has developed unique lipid metabolism, but the mechanisms remain unclear. Here, the significantly increased body weight of Arctic foxes was consistent with the significantly increased serum very-low-density lipoprotein (VLDL), and the 40% crude fat diet further increased the Arctic fox body weight. The enhanced body weight gain stems primarily from increased subcutaneous adipose tissue accumulation. The adipose triacylglycerol and phosphatidylethanolamine were significantly greater in Arctic foxes. The adipose fatty-acid synthase content was significantly lower in Arctic foxes, highlighting the main role of exogenous fatty-acids in fat accumulation. Considering the same diet, liver-derived fat dominates adipose expansion in Arctic foxes. Liver transcriptome analysis revealed greater fat and VLDL synthesis in Arctic foxes, consistent with the greater VLDL. Glucose homeostasis wasn't impacted in Arctic foxes. And the free fatty-acids in adipose, which promote insulin resistance, also did not differ between groups. However, the hepatic glycogen was greater in Arctic foxes and transcriptome analysis revealed upregulated glycogen synthesis, improving glucose homeostasis. These results suggest that the superior fat accumulation capacity and distinct characteristics of hepatic and adipose lipid and glucose metabolism facilitate glucose homeostasis and massive fat accumulation in Arctic foxes.
Collapse
Affiliation(s)
- Yuhang Zhu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Yuan Yuan
- School of Ecology and Environment, Northwestern Polytechnical University, Xi’an, China
| | - Huazhe Si
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Songze Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Fei Zhao
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Ruina Mu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Zihan Lin
- College of Plant Protection, Jilin Agricultural University, Changchun, China
| | - Xiaoxu Wang
- Department of Special Animal Nutrition and Feed Science, Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, China
| | - Qiang Qiu
- School of Ecology and Environment, Northwestern Polytechnical University, Xi’an, China
| | - Chao Xu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Lele Ji
- National Demonstration Center for Experimental Preclinical Medicine Education, The Fourth Military Medical University, Xi'an, China
| | - Zhipeng Li
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, China
- Key Lab of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
38
|
Mathiowetz AJ, Meymand ES, Deol KK, Parlakgül G, Lange M, Pang SP, Roberts MA, Torres EF, Jorgens DM, Zalpuri R, Kang M, Boone C, Zhang Y, Morgens DW, Tso E, Zhou Y, Talukdar S, Levine TP, Ku G, Arruda AP, Olzmann JA. CLCC1 promotes hepatic neutral lipid flux and nuclear pore complex assembly. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597858. [PMID: 38895340 PMCID: PMC11185754 DOI: 10.1101/2024.06.07.597858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Imbalances in lipid storage and secretion lead to the accumulation of hepatocyte lipid droplets (LDs) (i.e., hepatic steatosis). Our understanding of the mechanisms that govern the channeling of hepatocyte neutral lipids towards cytosolic LDs or secreted lipoproteins remains incomplete. Here, we performed a series of CRISPR-Cas9 screens under different metabolic states to uncover mechanisms of hepatic neutral lipid flux. Clustering of chemical-genetic interactions identified CLIC-like chloride channel 1 (CLCC1) as a critical regulator of neutral lipid storage and secretion. Loss of CLCC1 resulted in the buildup of large LDs in hepatoma cells and knockout in mice caused liver steatosis. Remarkably, the LDs are in the lumen of the ER and exhibit properties of lipoproteins, indicating a profound shift in neutral lipid flux. Finally, remote homology searches identified a domain in CLCC1 that is homologous to yeast Brl1p and Brr6p, factors that promote the fusion of the inner and outer nuclear envelopes during nuclear pore complex assembly. Loss of CLCC1 lead to extensive nuclear membrane herniations, consistent with impaired nuclear pore complex assembly. Thus, we identify CLCC1 as the human Brl1p/Brr6p homolog and propose that CLCC1-mediated membrane remodeling promotes hepatic neutral lipid flux and nuclear pore complex assembly.
Collapse
Affiliation(s)
- Alyssa J. Mathiowetz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily S. Meymand
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kirandeep K. Deol
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Güneş Parlakgül
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mike Lange
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Stephany P. Pang
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Melissa A. Roberts
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily F. Torres
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Danielle M. Jorgens
- Electron Microscope Laboratory, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Reena Zalpuri
- Electron Microscope Laboratory, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Misun Kang
- Electron Microscope Laboratory, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Casadora Boone
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Yaohuan Zhang
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David W. Morgens
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily Tso
- Merck & Co., Inc., South San Francisco, CA 94080, USA
| | | | | | - Tim P. Levine
- University College London InsYtute of Ophthalmology, Bath Street London, EC1V 9EL, UK
| | - Gregory Ku
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
- Department of Medicine, Division of Endocrinology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ana Paula Arruda
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - James A. Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of NutriYonal Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| |
Collapse
|
39
|
Qiu M, Yang L, Jiang Z, Chen Y, Liu Q, Wang X, Qu W. Mixed exposure to haloacetaldehyde disinfection by-products exacerbates lipid aggregation in the liver of mice. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 350:123971. [PMID: 38641033 DOI: 10.1016/j.envpol.2024.123971] [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: 02/29/2024] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
Haloacetaldehyde disinfection by-products (HAL-DBPs) are among the top three unregulated DBPs found in drinking water. The cytotoxicity and genotoxicity of HALs are much higher than that of the regulated trihalomethanes and haloacetic acids. Previous studies have mainly focused on the toxic effects of single HAL, with few examining the toxic effects of mixed exposures to HALs. The study aimed to observe the effects of mixed exposures of 1∼1000X the realistic level of HALs on the hepatotoxicity and lipid metabolism of C57BL/6J mice, based on the component and concentration of HALs detected in the finished water of Shanghai. Exposure to realistic levels of HALs led to a significant increase in phosphorated acetyl CoA carboxylase 1 (p-ACC1) in the hepatic de novo lipogenesis (DNL) pathway. Additionally, exposure to 100X realistic levels of HALs resulted in significant alterations to key enzymes of DNL pathway, including ACC1, fatty acid synthase (FAS), and diacylglycerol acyltransferase 2 (DGAT2), as well as key proteins of lipid disposal such as carnitine palmitoyltransferase 1 (CPT-1) and peroxisome proliferator activated receptor α (PPARα). Exposure to 1000X realistic levels of HALs significantly increased hepatic and serum triglyceride levels, as well as total cholesterol, low-density lipoprotein, alanine aminotransferase, aspartate transaminase, alkaline phosphatase, and lactate dehydrogenase levels, significantly decreased high-density lipoprotein. Meanwhile, histopathological analysis demonstrated that HALs exacerbated tissue vacuolization and inflammatory cell infiltration in mice livers, which showed the typical phenotypes of non-alcoholic fatty liver disease (NAFLD). These results suggested that the HALs mixture is a critical risk factor for NAFLD and is significantly highly toxic to C57BL/6J mice.
Collapse
Affiliation(s)
- Meiyue Qiu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Lili Yang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Zhiqiang Jiang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Yu Chen
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Qinxin Liu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Xia Wang
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| | - Weidong Qu
- Center for Water and Health, Key Laboratory of the Public Health Safety, Ministry of Education, Department of Environmental Health, School of Public Health, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
40
|
Malis Y, Armoza-Eilat S, Nevo-Yassaf I, Dukhovny A, Sklan EH, Hirschberg K. Rab1b facilitates lipid droplet growth by ER-to-lipid droplet targeting of DGAT2. SCIENCE ADVANCES 2024; 10:eade7753. [PMID: 38809969 PMCID: PMC11135398 DOI: 10.1126/sciadv.ade7753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 04/16/2024] [Indexed: 05/31/2024]
Abstract
Lipid droplets (LDs) comprise a triglyceride core surrounded by a lipid monolayer enriched with proteins, many of which function in LD homeostasis. How proteins are targeted to the growing LD is still unclear. Rab1b, a GTPase regulating secretory transport, was recently associated with targeting proteins to LDs in a Drosophila RNAi screen. LD formation was prevented in human hepatoma cells overexpressing dominant-negative Rab1b. We thus hypothesized that Rab1b recruits lipid-synthesizing enzymes, facilitating LD growth. Here, FRET between diacylglycerol acyltransferase 2 (DGAT2) and Rab1b and activity mutants of the latter demonstrated that Rab1b promotes DGAT2 ER to the LD surface redistribution. Last, alterations in LD metabolism and DGAT2 redistribution, consistent with Rab1b activity, were caused by mutations in the Rab1b-GTPase activating protein TBC1D20 in Warburg Micro syndrome (WARBM) model mice fibroblasts. These data contribute to our understanding of the mechanism of Rab1b in LD homeostasis and WARBM, a devastating autosomal-recessive disorder caused by mutations in TBC1D20.
Collapse
Affiliation(s)
- Yehonathan Malis
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shir Armoza-Eilat
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Inbar Nevo-Yassaf
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anna Dukhovny
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ella H. Sklan
- Department of Clinical Microbiology and Immunology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Koret Hirschberg
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
41
|
Graff J, Schneiter R. FIT2 proteins and lipid droplet emergence, an interplay between phospholipid synthesis, surface tension, and membrane curvature. Front Cell Dev Biol 2024; 12:1422032. [PMID: 38872930 PMCID: PMC11169642 DOI: 10.3389/fcell.2024.1422032] [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: 04/23/2024] [Accepted: 05/06/2024] [Indexed: 06/15/2024] Open
Abstract
Lipid droplets (LDs) serve as intracellular compartments primarily dedicated to the storage of metabolic energy in the form of neutral lipids. The processes that regulate and control LD biogenesis are being studied extensively and are gaining significance due to their implications in major metabolic disorders, including type 2 diabetes and obesity. A protein of particular interest is Fat storage-Inducing Transmembrane 2 (FIT2), which affects the emergence step of LD biogenesis. Instead of properly emerging towards the cytosol, LDs in FIT2-deficient cells remain embedded within the membrane of the endoplasmic reticulum (ER). In vitro studies revealed the ability of FIT2 to bind both di- and triacylglycerol (DAG/TAG), key players in lipid storage, and its activity to cleave acyl-CoA. However, the translation of these in vitro functions to the observed embedding of LDs in FIT2 deficient cells remains to be established. To understand the role of FIT2 in vivo, we discuss the parameters that affect LD emergence. Our focus centers on the role that membrane curvature and surface tension play in LD emergence, as well as the impact that the lipid composition exerts on these key parameters. In addition, we discuss hypotheses on how FIT2 could function locally to modulate lipids at sites of LD emergence.
Collapse
Affiliation(s)
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
42
|
Han HK, Mukherjee S, Park SY, Lee JH, Lee EH, Kim S, Lee YH, Song DK, Lee S, Bae JH, Im SS. Regulation of Betaine Homocysteine Methyltransferase by Liver Receptor Homolog-1 in the Methionine Cycle. Mol Cell Biol 2024; 44:245-258. [PMID: 38804232 PMCID: PMC11204035 DOI: 10.1080/10985549.2024.2354821] [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: 11/08/2023] [Accepted: 05/08/2024] [Indexed: 05/29/2024] Open
Abstract
Betaine-homocysteine S-methyltransferase (BHMT) is one of the most abundant proteins in the liver and regulates homocysteine metabolism. However, the molecular mechanisms underlying Bhmt transcription have not yet been elucidated. This study aimed to assess the molecular mechanisms underlying Bhmt transcription and the effect of BHMT deficiency on metabolic functions in the liver mediated by liver receptor homolog-1 (LRH-1). During fasting, both Bhmt and Lrh-1 expression increased in the liver of Lrh-1f/f mice; however, Bhmt expression was decreased in LRH-1 liver specific knockout mice. Promoter activity analysis confirmed that LRH-1 binds to a specific site in the Bhmt promoter region. LRH-1 deficiency was associated with elevated production of reactive oxygen species (ROS), lipid peroxidation, and mitochondrial stress in hepatocytes, contributing to hepatic triglyceride (TG) accumulation. In conclusion, this study suggests that the absence of an LRH-1-mediated decrease in Bhmt expression promotes TG accumulation by increasing ROS levels and inducing mitochondrial stress. Therefore, LRH-1 deficiency not only leads to excess ROS production and mitochondrial stress in hepatocytes, but also disrupts the methionine cycle. Understanding these regulatory pathways may pave the way for novel therapeutic interventions against metabolic disorders associated with hepatic lipid accumulation.
Collapse
Affiliation(s)
- Hee-Kyung Han
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Sulagna Mukherjee
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Soo-Young Park
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Jae-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Eun-Ho Lee
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Suji Kim
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Yun Han Lee
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Dae-Kyu Song
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Sooyeun Lee
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Jae-Hoon Bae
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Seung-Soon Im
- Department of Physiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| |
Collapse
|
43
|
Ariyoshi K, Nishiyama K, Kato Y, Mi X, Ito T, Azuma YT, Nishimura A, Nishida M. Inhibition of Drp1-Filamin Protein Complex Prevents Hepatic Lipid Droplet Accumulation by Increasing Mitochondria-Lipid Droplet Contact. Int J Mol Sci 2024; 25:5446. [PMID: 38791484 PMCID: PMC11122359 DOI: 10.3390/ijms25105446] [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: 04/14/2024] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Lipid droplet (LD) accumulation in hepatocytes is one of the major symptoms associated with fatty liver disease. Mitochondria play a key role in catabolizing fatty acids for energy production through β-oxidation. The interplay between mitochondria and LD assumes a crucial role in lipid metabolism, while it is obscure how mitochondrial morphology affects systemic lipid metabolism in the liver. We previously reported that cilnidipine, an already existing anti-hypertensive drug, can prevent pathological mitochondrial fission by inhibiting protein-protein interaction between dynamin-related protein 1 (Drp1) and filamin, an actin-binding protein. Here, we found that cilnidipine and its new dihydropyridine (DHP) derivative, 1,4-DHP, which lacks Ca2+ channel-blocking action of cilnidipine, prevent the palmitic acid-induced Drp1-filamin interaction, LD accumulation and cytotoxicity of human hepatic HepG2 cells. Cilnidipine and 1,4-DHP also suppressed the LD accumulation accompanied by reducing mitochondrial contact with LD in obese model and high-fat diet-fed mouse livers. These results propose that targeting the Drp1-filamin interaction become a new strategy for the prevention or treatment of fatty liver disease.
Collapse
Affiliation(s)
- Kohei Ariyoshi
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
| | - Kazuhiro Nishiyama
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
- Laboratory of Prophylactic Pharmacology, Osaka Metropolitan University Graduate School of Veterinary Science, Osaka 598-8531, Japan;
| | - Yuri Kato
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
| | - Xinya Mi
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
| | - Tomoya Ito
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
| | - Yasu-Taka Azuma
- Laboratory of Prophylactic Pharmacology, Osaka Metropolitan University Graduate School of Veterinary Science, Osaka 598-8531, Japan;
| | - Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| | - Motohiro Nishida
- Department of Physiology, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan; (K.A.); (K.N.); (Y.K.); (X.M.); (T.I.)
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan;
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences (NINS), Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Okazaki 444-8787, Japan
| |
Collapse
|
44
|
Maurotti S, Geirola N, Frosina M, Mirarchi A, Scionti F, Mare R, Montalcini T, Pujia A, Tirinato L. Exploring the impact of lipid droplets on the evolution and progress of hepatocarcinoma. Front Cell Dev Biol 2024; 12:1404006. [PMID: 38818407 PMCID: PMC11137176 DOI: 10.3389/fcell.2024.1404006] [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: 03/20/2024] [Accepted: 04/29/2024] [Indexed: 06/01/2024] Open
Abstract
Over the past 10 years, the biological role of lipid droplets (LDs) has gained significant attention in the context of both physiological and pathological conditions. Considerable progress has been made in elucidating key aspects of these organelles, yet much remains to be accomplished to fully comprehend the myriad functions they serve in the progression of hepatic tumors. Our current perception is that LDs are complex and active structures managed by a distinct set of cellular processes. This understanding represents a significant paradigm shift from earlier perspectives. In this review, we aim to recapitulate the function of LDs within the liver, highlighting their pivotal role in the pathogenesis of metabolic dysfunction-associated steatotic liver disease (MASLD) (Hsu and Loomba, 2024) and their contribution to the progression towards more advanced pathological stages up to hepatocellular carcinoma (HC) (Farese and Walther, 2009). We are aware of the molecular complexity and changes occurring in the neoplastic evolution of the liver. Our attempt, however, is to summarize the most important and recent roles of LDs across both healthy and all pathological liver states, up to hepatocarcinoma. For more detailed insights, we direct readers to some of the many excellent reviews already available in the literature (Gluchowski et al., 2017; Hu et al., 2020; Seebacher et al., 2020; Paul et al., 2022).
Collapse
Affiliation(s)
- Samantha Maurotti
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Nadia Geirola
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Miriam Frosina
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Angela Mirarchi
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Francesca Scionti
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Rosario Mare
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Tiziana Montalcini
- Department of Clinical and Experimental Medicine, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Arturo Pujia
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| | - Luca Tirinato
- Department of Medical and Surgical Sciences, University “Magna Græcia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
45
|
Jia W, Zhong L, Ren Q, Teng D, Gong L, Dong H, Li J, Wang C, He YX, Yang J. Microcystin-RR promote lipid accumulation through CD36 mediated signal pathway and fatty acid uptake in HepG2 cells. ENVIRONMENTAL RESEARCH 2024; 249:118402. [PMID: 38309560 DOI: 10.1016/j.envres.2024.118402] [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: 11/20/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/05/2024]
Abstract
Microcystins (MC)-RR is a significant analogue of MC-LR, which has been identified as a hepatotoxin capable of influencing lipid metabolism and promoting the progression of liver-related metabolic diseases. However, the toxicity and biological function of MC-RR are still not well understood. In this study, the toxic effects and its role in lipid metabolism of MC-RR were investigated in hepatoblastoma cells (HepG2cells). The results demonstrated that MC-RR dose-dependently reduced cell viability and induced apoptosis. Additionally, even at low concentrations, MC-RR promoted lipid accumulation through up-regulating levels of triglyceride, total cholesterol, phosphatidylcholines and phosphatidylethaolamine in HepG2 cells, with no impact on cell viability. Proteomics and transcriptomics analysis further revealed significant alterations in the protein and gene expression profiles in HepG2 cells treated with MC-RR. Bioinformatic analysis, along with subsequent validation, indicated the upregulation of CD36 and activation of the AMPK and PI3K/AKT/mTOR in response to MC-RR exposure. Finally, knockdown of CD36 markedly ameliorated MC-RR-induced lipid accumulation in HepG2 cells. These findings collectively suggest that MC-RR promotes lipid accumulation in HepG2 cells through CD36-mediated signal pathway and fatty acid uptake. Our findings provide new insights into the hepatotoxic mechanism of MC-RR.
Collapse
Affiliation(s)
- Wenjuan Jia
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| | - Lin Zhong
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| | - Da Teng
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Lei Gong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Haibin Dong
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Jun Li
- School of Basic Medical Sciences, Qingdao University, Qingdao, 266071, China; Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Chunxiao Wang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China
| | - Yong-Xing He
- Ministry of Education Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Jun Yang
- Department of Cardiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai, 264000, China.
| |
Collapse
|
46
|
Wang Q, Xiong J, He Y, He J, Cai M, Luo Z, Zhang T, Zhou X. Effect of L-arabinose and lactulose combined with Lactobacillus plantarum on obesity induced by a high-fat diet in mice. Food Funct 2024; 15:5073-5087. [PMID: 38656276 DOI: 10.1039/d4fo00369a] [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
L-Arabinose, lactulose, and Lactobacillus plantarum (L. plantarum) have been reported to have glucolipid-lowering effects. Here, the effects of L-arabinose and lactulose combined with L. plantarum on obesity traits were investigated. According to the experimental results, the combination of L-arabinose, lactulose, and L. plantarum was more effective at reducing body weight, regulating glucolipid metabolism, and improving insulin resistance. Besides, this combination showed immunomodulatory activity by adjusting the T lymphocyte subsets and reduced the immune-related cytokine production. Moreover, it improved the gut barrier, ameliorated the disorder of gut microbiota, and upregulated the levels of SCFAs. More importantly, the AL group, LP group, and ALLP group showed different regulatory effects on the abundance of Bifidobacterium and Lactobacillus due to the presence of lactulose and L. plantarum. These findings elucidate that the combination of L-arabinose, lactulose, and L. plantarum constitutes a new synbiotic combination to control obesity by modulating glucolipid metabolism, immunomodulatory activity, inflammation, gut barrier, gut microbiota and production of SCFAs.
Collapse
Affiliation(s)
- Qiong Wang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Jialu Xiong
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Yalun He
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Juncheng He
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Miaomiao Cai
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Zexian Luo
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Tongcun Zhang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| | - Xiang Zhou
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China.
| |
Collapse
|
47
|
Plötz T, Lenzen S. Mechanisms of lipotoxicity-induced dysfunction and death of human pancreatic beta cells under obesity and type 2 diabetes conditions. Obes Rev 2024; 25:e13703. [PMID: 38327101 DOI: 10.1111/obr.13703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 12/06/2023] [Accepted: 12/20/2023] [Indexed: 02/09/2024]
Abstract
The term "pancreatic beta-cell lipotoxicity" refers to the detrimental effects of free fatty acids (FFAs) on a wide variety of cellular functions. Basic research in the field has primarily analyzed the effects of palmitic acid and oleic acid. The focus on these two physiological FFAs, however, ignores differences in chain length and degree of saturation. In order to gain a comprehensive understanding of the lipotoxic mechanisms, a wide range of structurally related FFAs should be investigated. Structure-activity relationship analyses of FFAs in the human EndoC-βH1 beta-cell line have provided a deep insight into the mechanisms of beta-cell lipotoxicity. This review focuses on the effects of a wide range of FFAs with crucial structural determinants for the development of lipotoxicity in human beta cells and documents an association between increased triglyceride stores in obesity and in type 2 diabetes.
Collapse
Affiliation(s)
- Thomas Plötz
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany
| |
Collapse
|
48
|
Sapia J, Vanni S. Molecular dynamics simulations of intracellular lipid droplets: a new tool in the toolbox. FEBS Lett 2024; 598:1143-1153. [PMID: 38627196 DOI: 10.1002/1873-3468.14879] [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: 11/17/2023] [Revised: 03/14/2024] [Accepted: 03/25/2024] [Indexed: 05/25/2024]
Abstract
Lipid droplets (LDs) are ubiquitous intracellular organelles with a central role in multiple lipid metabolic pathways. However, identifying correlations between their structural properties and their biological activity has proved challenging, owing to their unique physicochemical properties as compared with other cellular membranes. In recent years, molecular dynamics (MD) simulations, a computational methodology allowing the accurate description of molecular assemblies down to their individual components, have been demonstrated to be a useful and powerful approach for studying LD structural and dynamical properties. In this short review, we attempt to highlight, as comprehensively as possible, how MD simulations have contributed to our current understanding of multiple molecular mechanisms involved in LD biology.
Collapse
Affiliation(s)
- Jennifer Sapia
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Stefano Vanni
- Department of Biology, University of Fribourg, Fribourg, Switzerland
- Université Côte d'Azur, Inserm, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
- Swiss National Center for Competence in Research (NCCR) Bio-inspired Materials, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
49
|
Dibwe DF, Kitayama E, Oba S, Takeishi N, Chiba H, Hui SP. Inhibition of Lipid Accumulation and Oxidation in Hepatocytes by Bioactive Bean Extracts. Antioxidants (Basel) 2024; 13:513. [PMID: 38790618 PMCID: PMC11118026 DOI: 10.3390/antiox13050513] [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: 03/07/2024] [Revised: 04/13/2024] [Accepted: 04/14/2024] [Indexed: 05/26/2024] Open
Abstract
During our search for natural resources that can inhibit lipid droplet accumulation (LDA) and potentially prevent metabolic dysfunction-associated fatty liver disease (MAFLD) and its progressive stages, such as metabolic dysfunction-associated steatohepatitis (MASH), eight bean extracts (BE1-BE8) were tested for their ability to inhibit lipid accumulation and oxidation in hepatocytes. Substantial inhibitory effects on LDA with bean extracts (BEs) BE2, BE4, BE5, and BE8 were demonstrated. An advanced lipidomic approach was used to quantify the accumulation and inhibition of intracellular triacylglycerol (TAG) and its oxidized species, TAG hydroperoxide (TGOOH), in hepatocytes under fatty acid-loading conditions. The results show that the antioxidants BE2 and BE8 are potential candidates for regulating TAG and TGOOH accumulation in fatty acid-induced lipid droplets (LDs). This study suggests that bean-based foods inhibit LDs formation by decreasing intracellular lipids and lipid hydroperoxides in the hepatocytes. The metabolic profiling of BEs revealed that BE2 and BE8 contained polyphenolic compounds. These may be potential resources for the development of functional foods and drug discovery targeting MAFLD/MASH.
Collapse
Affiliation(s)
- Dya Fita Dibwe
- Faculty of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-Ku, Sapporo 060-0812, Japan;
| | - Emi Kitayama
- Graduate School of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-Ku, Sapporo 060-0812, Japan; (E.K.); (S.O.); (N.T.)
| | - Saki Oba
- Graduate School of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-Ku, Sapporo 060-0812, Japan; (E.K.); (S.O.); (N.T.)
| | - Nire Takeishi
- Graduate School of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-Ku, Sapporo 060-0812, Japan; (E.K.); (S.O.); (N.T.)
| | - Hitoshi Chiba
- Department of Nutrition, Sapporo University of Health Sciences, Nakanuma Nishi-4-3-1-15, Higashi-Ku, Sapporo 007-0894, Japan;
| | - Shu-Ping Hui
- Faculty of Health Sciences, Hokkaido University, Kita-12, Nishi-5, Kita-Ku, Sapporo 060-0812, Japan;
| |
Collapse
|
50
|
Qin S, Wang X, Jiang Y. Dual-state emission, mechanofluorochromism, and lipid droplet imaging of asymmetric D-π-A-D'-type triads. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 311:124014. [PMID: 38354679 DOI: 10.1016/j.saa.2024.124014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/15/2024] [Accepted: 02/06/2024] [Indexed: 02/16/2024]
Abstract
Dual-state emission (DSE) is an emerging phenomenon wherein organic luminescent molecules display bright emissions in both molecularly isolated and packed states, addressing the challenge associated with the traditional paradigm of dyes with mono-state emission. This study presents the design and synthesis of two unsymmetrical triads, TPCA and TPCT, featuring a D-π-A-D' electronic structure by integrating phenothiazines, triphenylamines, and cyanostilbene. Photophysical assessments reveal that both molecules serve as robust DSEgens, exhibiting strong emissions in both solution and solid phases. TPCA displays ΦTHF 53.2% and Φsolids 43.2%, while TPCT exhibits ΦTHF 49.6% and Φsolids 37.5%. However, due to differences in molecular conformation and packing, they diverge in solid-state emission wavelengths and mechanofluorochromic behavior. In the solid state, TPCA emits strong red fluorescence, contrasting with TPCT, which emits orange fluorescence. Furthermore, TPCA demonstrates significant mechanofluorochromism (MFC), shifting from yellow to yellow-red upon mechanical grinding, while TPCT exhibits negligible MFC owing to conformational distinctions. As robust and low-toxic bioimaging agents, both TPCA and TPCT prove highly effective for lipid-droplet imaging studies. This research contributes valuable insights to the evolving field of DSE materials, elucidating the promising applications and mechanisms governing their versatile emission behaviors.
Collapse
Affiliation(s)
- Sihao Qin
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, the First Hospital of Jilin University, Changchun 130021, China.
| | - Xueyao Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun 130021, China
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration & Transplantation of the Ministry of Education, Genetic Diagnosis Center, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|