51
|
Neuditschko B, King AP, Huang Z, Janker L, Bileck A, Borutzki Y, Marker SC, Gerner C, Wilson JJ, Meier‐Menches SM. An Anticancer Rhenium Tricarbonyl Targets Fe-S Cluster Biogenesis in Ovarian Cancer Cells. Angew Chem Int Ed Engl 2022; 61:e202209136. [PMID: 36004624 PMCID: PMC9827826 DOI: 10.1002/anie.202209136] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Indexed: 01/12/2023]
Abstract
Target identification remains a critical challenge in inorganic drug discovery to deconvolute potential polypharmacology. Herein, we describe an improved approach to prioritize candidate protein targets based on a combination of dose-dependent chemoproteomics and treatment effects in living cancer cells for the rhenium tricarbonyl compound TRIP. Chemoproteomics revealed 89 distinct dose-dependent targets with concentrations of competitive saturation between 0.1 and 32 μM despite the broad proteotoxic effects of TRIP. Target-response networks revealed two highly probable targets of which the Fe-S cluster biogenesis factor NUBP2 was competitively saturated by free TRIP at nanomolar concentrations. Importantly, TRIP treatment led to a down-regulation of Fe-S cluster containing proteins and upregulated ferritin. Fe-S cluster depletion was further verified by assessing mitochondrial bioenergetics. Consequently, TRIP emerges as a first-in-class modulator of the scaffold protein NUBP2, which disturbs Fe-S cluster biogenesis at sub-cytotoxic concentrations in ovarian cancer cells.
Collapse
Affiliation(s)
- Benjamin Neuditschko
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Present address: Institute Krems BioanalyticsIMC University of Applied Sciences Krems3500KremsAustria
| | - A. Paden King
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA,Present address: Chemical Biology LaboratoryCenter for Cancer ResearchNational Cancer InstituteFrederickMD 21702USA
| | - Zhouyang Huang
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA
| | - Lukas Janker
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Andrea Bileck
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Yasmin Borutzki
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria
| | - Sierra C. Marker
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA,Present address: Chemical Biology LaboratoryCenter for Cancer ResearchNational Cancer InstituteFrederickMD 21702USA
| | - Christopher Gerner
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| | - Justin J. Wilson
- Department of Chemistry and Chemical BiologyCornell UniversityIthacaNY 14853USA
| | - Samuel M. Meier‐Menches
- Department of Analytical ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Institute of Inorganic ChemistryFaculty of ChemistryUniversity of Vienna1090ViennaAustria,Joint Metabolome FacilityUniversity of Vienna and Medical University Vienna1090ViennaAustria
| |
Collapse
|
52
|
Volkmar N, Gawden‐Bone CM, Williamson JC, Nixon‐Abell J, West JA, St George‐Hyslop PH, Kaser A, Lehner PJ. Regulation of membrane fluidity by RNF145-triggered degradation of the lipid hydrolase ADIPOR2. EMBO J 2022; 41:e110777. [PMID: 35993436 PMCID: PMC9531299 DOI: 10.15252/embj.2022110777] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/19/2022] Open
Abstract
The regulation of membrane lipid composition is critical for cellular homeostasis. Cells are particularly sensitive to phospholipid saturation, with increased saturation causing membrane rigidification and lipotoxicity. How mammalian cells sense membrane lipid composition and reverse fatty acid (FA)-induced membrane rigidification is poorly understood. Here we systematically identify proteins that differ between mammalian cells fed saturated versus unsaturated FAs. The most differentially expressed proteins were two ER-resident polytopic membrane proteins: the E3 ubiquitin ligase RNF145 and the lipid hydrolase ADIPOR2. In unsaturated lipid membranes, RNF145 is stable, promoting its lipid-sensitive interaction, ubiquitination and degradation of ADIPOR2. When membranes become enriched in saturated FAs, RNF145 is rapidly auto-ubiquitinated and degraded, stabilising ADIPOR2, whose hydrolase activity restores lipid homeostasis and prevents lipotoxicity. We therefore identify RNF145 as a FA-responsive ubiquitin ligase which, together with ADIPOR2, defines an autoregulatory pathway that controls cellular membrane lipid homeostasis and prevents acute lipotoxic stress.
Collapse
Affiliation(s)
- Norbert Volkmar
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
- Present address:
Institute for Molecular Systems Biology (IMSB)ETH ZürichZürichSwitzerland
| | - Christian M Gawden‐Bone
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | - James C Williamson
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | | | - James A West
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | | | - Arthur Kaser
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| | - Paul J Lehner
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical CentreUniversity of CambridgeCambridgeUK
| |
Collapse
|
53
|
Leikin-Frenkel A, Cohen H, Keshet R, Shnerb-GanOr R, Kandel-Kfir M, Harari A, Hollander KS, Shaish A, Harats D, Kamari Y. The effect of α-linolenic acid enrichment in perinatal diets in preventing high fat diet-induced SCD1 increased activity and lipid disarray in adult offspring of low density lipoprotein receptor knockout (LDLRKO) mice. Prostaglandins Leukot Essent Fatty Acids 2022; 184:102475. [PMID: 35940045 DOI: 10.1016/j.plefa.2022.102475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/29/2022]
Abstract
The present study examined the effects of maternal perinatal dietary ALA enrichment on the high fat diet (HFD)-induced lipid disarray in the adult offspring of low density lipoprotein receptor knock-out (LDLRKO) mice. Female LDLRKO mice received, during pregnancy and lactation, isocaloric diets with either corn oil, RD, or flax oil, ALA. The weaning offspring was given a regular chow diet for a washout period of eight weeks, which was followed by HFD for eight weeks. Plasma and liver lipids and SCD1 activity were then analyzed. The HFD-fed RD adult offspring had substantially higher plasma cholesterol levels than the HFD-fed ALA offspring (15.7 versus 9.7 mmole/l, p<0.00001) and non-alcoholic fatty liver disease (NAFLD) (65.0 versus 23.9 mg/g lipids, p<0.00001). Liver lipids oleic acid (OA) content and monounsaturated to saturated fatty acids (MUFA/SAT) ratio, were two times lower in RD compared to ALA (p<0.0001). The threefold HFD-induced SCD1 raised activity (p<0.00001), and OA produced from SA, observed in RD adult offspring were prevented by perinatal ALA. In conclusion, the resilience of SCD1 to HFD- induced increased activity may account for the beneficial effects of perinatal ALA dietary enrichment in preventing NAFLD and hypercholesterolemia from occurring in adult LDLRKO offspring mice.
Collapse
Affiliation(s)
- A Leikin-Frenkel
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel.
| | - H Cohen
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | - R Keshet
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - R Shnerb-GanOr
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - M Kandel-Kfir
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - A Harari
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - K S Hollander
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | - A Shaish
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Achva Academic College, Israel
| | - D Harats
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| | - Y Kamari
- The Bert W. Strassburger Metabolic Center, Sheba Medical Center, Tel-Hashomer, 5265601, Israel; Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, 69978, Israel
| |
Collapse
|
54
|
Yang L, Li JZ, Li MR. Progress in research of lipogenesis inhibitors for treatment of nonalcoholic fatty liver disease. Shijie Huaren Xiaohua Zazhi 2022; 30:735-742. [DOI: 10.11569/wcjd.v30.i16.735] [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] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the main cause of chronic liver disease. At present, the main clinical treatment for NAFLD is diet adjustment, exercise, and weight loss, but the effect is poor, and there is still a lack of recognized drugs with significant efficacy in NAFLD. In recent years, with the in-depth study of the pathogenesis of NAFLD, it has been found that the core enzymes that inhibit intrahepatic de novo lipogenesis (DNL), including citrate/isocitrate carrier (CIC), ATP-citrate lyase (ACLY), acetyl-CoA carboxylase (ACC), fatty acid synthase (FASN), and stearoyl-CoA desaturase 1 (SCD1), can improve hepatic steatosis and provide a new method for the treatment of NAFLD. This article reviews the research progress of five different types of lipogenesis inhibitors for treatment of NAFLD.
Collapse
Affiliation(s)
- Liu Yang
- Department of Infectious Diseases, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Jin-Zhong Li
- Department of Infectious Diseases, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| | - Min-Ran Li
- Department of Infectious Diseases, The First Affiliated Hospital of Jinan University, Guangzhou 510630, Guangdong Province, China
| |
Collapse
|
55
|
Neuditschko B, King AP, Huang Z, Janker L, Bileck A, Borutzki Y, Marker SC, Gerner C, Wilson JJ, Meier-Menches SM. An Anticancer Rhenium Tricarbonyl Targets Fe‐S Cluster Biogenesis in Ovarian Cancer Cells. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202209136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Benjamin Neuditschko
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - A. Paden King
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Zhouyang Huang
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Lukas Janker
- University of Vienna Faculty of Chemistry: Universitat Wien Fakultat fur Chemie Department of Analytical Chemistry AUSTRIA
| | - Andrea Bileck
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - Yasmin Borutzki
- University of Vienna: Universitat Wien Institute of Inorganic Chemistry AUSTRIA
| | - Sierra C. Marker
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Christopher Gerner
- University of Vienna: Universitat Wien Department of Analytical Chemistry AUSTRIA
| | - Justin J. Wilson
- Cornell University Department of Chemistry and Chemical Biology UNITED STATES
| | - Samuel M. Meier-Menches
- University of Vienna: Universitat Wien Department of Analytical Chemistry Waehringer Str. 38 1090 Vienna AUSTRIA
| |
Collapse
|
56
|
Yoon CH, Ryu JS, Ko JH, Kim YK, Oh JH, Chung JH, Oh JY. The Eyelid Meibomian Gland Deficiency in Fucosyltransferase 1 Knockout Mice. Int J Mol Sci 2022; 23:ijms23169464. [PMID: 36012728 PMCID: PMC9409428 DOI: 10.3390/ijms23169464] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
To investigate the effect of fucosyltransferase (FUT) 1-mediated fucosylation on meibomian glands (MG), we first confirmed that FUT1 and its fucosylated products were expressed in the eyelid, conjunctiva and skin in wild-type (WT) mice, whereas their mRNA and protein levels were downregulated in Fut1 knock-out (KO) mice. We then evaluated age-dependent changes in the total and acinar areas of MG, meibocyte differentiation, lipid synthesis, and eyelid inflammation and oxidative stress in Fut1 KO and WT mice. Results show that both the total and acinar areas of MG were smaller in Fut1 KO mice than in WT mice in all evaluated age groups. Meibocyte differentiation, lipid-producing capacities and the enzyme levels responsible for lipid synthesis were reduced in Fut1 KO mice, compared to WT controls. The levels of pro-inflammatory cytokines and oxidative-stress-related markers were elevated in the eyelids and MG of FUT1 KO mice. These findings demonstrate the physiologic function of FUT1-mediated fucosylation in MG development and function, and indicate its potential role in ocular surface homeostasis.
Collapse
Affiliation(s)
- Chang Ho Yoon
- Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Jin Suk Ryu
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Jung Hwa Ko
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Yeon Kyung Kim
- Department of Dermatology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Jang-Hee Oh
- Department of Dermatology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
| | - Joo Youn Oh
- Department of Ophthalmology, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul 03080, Korea
- Laboratory of Ocular Regenerative Medicine and Immunology, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea
- Correspondence: or ; Tel.: +82-2-2072-0027
| |
Collapse
|
57
|
Ferreira EV, João Júnior GC, Corrêa GSS, Kiefer C, Alencar SAS, Viana LH, Cavalheiro LF. Effects of organic Selenium- and Chromium-Enriched Diets on performance, carcass characteristics, lipid profile and fat quality of finishing pigs in different weight ranges. AN ACAD BRAS CIENC 2022; 94:e20200509. [PMID: 35946643 DOI: 10.1590/0001-3765202220200509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 05/11/2021] [Indexed: 11/21/2022] Open
Abstract
An experiment was conducted to evaluate the fatty acid profile of subcutaneous fat from barrowS of same genetic lineage supplemented with organic chromium and selenium initiated in different weight ranges in the finishing phase using 24 carcasses. Three different diets were used that represent the time when supplementation starts: control - without the inclusion of organic Cr and Se; CrSe70 - control with 500 g ton-1 of organic Cr and Se of 70 to 130 kg in body weight; and CrSe100 - control with inclusion of 500 g ton-1 of organic Cr and Se from 100 kg to 130 kg body weight. Performance, carcass characteristics, and lipid profile were evaluated. The data were submitted to analysis of variance, and with significant differences (p<0.05), the means were compared using the Tukey test. From 70 to 100 kg, control and CrSe70 animals consumed less feed than CrSe100. From 100 kg body weight, it reduced the C20:5n3 and C24:1n9 acids and increased the activity of the Δ-6 desaturase, elongase, Δ-5 desaturase enzymes in the supplemented animals. The moment when supplementation starts of organic chromium and selenium does not improve the performance and carcass characteristics, does not change the fatty acid profile, and does not improve the quality of the fat.
Collapse
Affiliation(s)
- Eduardo V Ferreira
- Universidade Federal de Mato Grosso, Faculdade de Agronomia e Zootecnia, Av. Fernando Corrêa da Costa, 2367, Boa Esperança, 78060-900 Cuiabá, MT, Brazil
| | - G C João Júnior
- Universidade Federal de Mato Grosso, Faculdade de Agronomia e Zootecnia, Av. Fernando Corrêa da Costa, 2367, Boa Esperança, 78060-900 Cuiabá, MT, Brazil
| | - Gerusa S S Corrêa
- Universidade Federal de Mato Grosso, Faculdade de Agronomia e Zootecnia, Av. Fernando Corrêa da Costa, 2367, Boa Esperança, 78060-900 Cuiabá, MT, Brazil
| | - Charles Kiefer
- Universidade Federal de Mato Grosso do Sul, Faculdade de Medicina Veterinária e Zootecnia, Av. Senador Filinto Müller, 2443, 79070-900 Campo Grande, MS, Brazil
| | - Stephan A S Alencar
- Universidade Federal de Mato Grosso do Sul, Instituto de Química, Av. Senador Filinto Müller, 1555, 79070-900 Campo Grande, MS, Brazil
| | - Luiz H Viana
- Universidade Federal de Mato Grosso do Sul, Instituto de Química, Av. Senador Filinto Müller, 1555, 79070-900 Campo Grande, MS, Brazil
| | - Leandro F Cavalheiro
- Universidade Federal de Mato Grosso do Sul, Instituto de Química, Av. Senador Filinto Müller, 1555, 79070-900 Campo Grande, MS, Brazil
| |
Collapse
|
58
|
Deng KQ, Huang X, Lei F, Zhang XJ, Zhang P, She ZG, Cai J, Ji YX, Li H. Role of hepatic lipid species in the progression of nonalcoholic fatty liver disease. Am J Physiol Cell Physiol 2022; 323:C630-C639. [PMID: 35759443 DOI: 10.1152/ajpcell.00123.2022] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has become the most common liver disease due to the global pandemic of metabolic diseases. Dysregulation of hepatic lipid metabolism plays a central role in the initiation and progression of NAFLD. With the advancement of lipidomics, an increasing number of lipid species and underlying mechanisms associating hepatic lipid components have been revealed. Therefore, the focus of this mini-review is to highlight the links between hepatic lipid species and their mechanisms mediating the pathogenesis of NAFLD. We first summarized the interplay between NAFLD and hepatic lipid disturbances. Next, we focused on reviewing the role of saturated fatty acids, cholesterol, oxidized phospholipids, and their respective intermediates in the pathogenesis of NAFLD. The mechanisms by which monounsaturated fatty acids and other pro-resolving mediators exert protective effects are also addressed. Finally, we further discussed the implication of different analysis approaches in lipidomic. Evolving insights into the pathophysiology of NAFLD will provide the opportunity for drug development.
Collapse
Affiliation(s)
- Ke-Qiong Deng
- Department of Cardiology, Center Hospital of Huanggang, Huanggang, China.,Huanggang Institute of Translation Medicine, Huanggang, China.,Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xuewei Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal, Wuhan University, Wuhan, China
| | - Fang Lei
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Peng Zhang
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Zhi-Gang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal, Wuhan University, Wuhan, China
| | - Jingjing Cai
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan-Xiao Ji
- Institute of Model Animal, Wuhan University, Wuhan, China.,School of Basic Medical Science, Wuhan University, Wuhan, China
| | - Hongliang Li
- Huanggang Institute of Translation Medicine, Huanggang, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China.,Institute of Model Animal, Wuhan University, Wuhan, China
| |
Collapse
|
59
|
Luo Y, Chen Q, Zou J, Fan J, Li Y, Luo Z. Chronic Intermittent Hypoxia Exposure Alternative to Exercise Alleviates High-Fat-Diet-Induced Obesity and Fatty Liver. Int J Mol Sci 2022; 23:ijms23095209. [PMID: 35563600 PMCID: PMC9104027 DOI: 10.3390/ijms23095209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/01/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity often concurs with nonalcoholic fatty liver disease (NAFLD), both of which are detrimental to human health. Thus far, exercise appears to be an effective treatment approach. However, its effects cannot last long and, moreover, it is difficult to achieve for many obese people. Thus, it is necessary to look into alternative remedies. The present study explored a noninvasive, easy, tolerable physical alternative. In our experiment, C57BL/6 mice were fed with a high-fat diet (HFD) to induce overweight/obesity and were exposed to 10% oxygen for one hour every day. We found that hypoxia exerted protective effects. First, it offset HFD-induced bodyweight gain and insulin resistance. Secondly, hypoxia reversed the HFD-induced enlargement of white and brown adipocytes and fatty liver, and protected liver function. Thirdly, HFD downregulated the expression of genes required for lipolysis and thermogenesis, such as UCP1, ADR3(beta3-adrenergic receptor), CPT1A, ATGL, PPARα, and PGC1α, M2 macrophage markers arginase and CD206 in the liver, and UCP1 and PPARγ in brown fat, while these molecules were upregulated by hypoxia. Furthermore, hypoxia induced the activation of AMPK, an energy sensing enzyme. Fourthly, our results showed that hypoxia increased serum levels of epinephrine. Indeed, the effects of hypoxia on bodyweight, fatty liver, and associated changes in gene expression ever tested were reproduced by injection of epinephrine and prevented by propranolol at varying degrees. Altogether, our data suggest that hypoxia triggers stress responses where epinephrine plays important roles. Therefore, our study sheds light on the hope to use hypoxia to treat the daunting disorders, obesity and NAFLD.
Collapse
Affiliation(s)
- Yunfei Luo
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Qiongfeng Chen
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Junrong Zou
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Jingjing Fan
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Yuanjun Li
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
| | - Zhijun Luo
- Jiangxi Provincial Key Laboratory of Tumor Pathogens and Molecular Pathology, Department of Pathophysiology, Schools of Basic Sciences, Nanchang University, Nanchang 330031, China; (Y.L.); (Q.C.); (J.Z.); (J.F.); (Y.L.)
- Queen Mary School, Nanchang University, Nanchang 330031, China
- Correspondence: ; Tel.: +86-158-7917-7010
| |
Collapse
|
60
|
Kang DH, Chung KY, Park BH, Kim UH, Jang SS, Smith ZK, Kim J. Effects of feeding high-energy diet on growth performance, blood parameters, and carcass traits in Hanwoo steers. Anim Biosci 2022; 35:1545-1555. [PMID: 35507848 PMCID: PMC9449379 DOI: 10.5713/ab.22.0014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/14/2022] [Indexed: 11/27/2022] Open
Abstract
Objective Our study aimed to investigate the effects of a 2% increase in dietary total digestible nutrients (TDN) value during the growing (7 to 12 mo of age) and fattening (13 to 30 mo of age) period of Hanwoo steers. Methods Two hundred and twenty Hanwoo steers were assigned to one of two treatments: i) a control group (basal TDN, BTDN, n = 111 steers, growing = 70.5%, early fattening = 71.0%, late fattening = 74.0%) or high TDN (HTDN, n = 109 steers, growing = 72.6%, early = 73.1%, late = 76.2%). Growth performance, carcass traits, blood parameters, and gene expression of longissimus dorsi (LD) (7, 18, and 30 mo) were quantified. Results Steers on the BTDN diets had increased (p≤0.02) DMI throughout the feeding trial compared to HTDN, but gain did not differ appreciably. A greater proportion of cattle in HTDN received Korean quality grade 1 (82%) or greater compared to BTDN (77%), while HTDN had a greater yield grade (29%) than BTDN (20%). Redness (a*) of LD muscle was improved (p = 0.021) in steers fed HTDN. Feeding the HTDN diet did not alter blood parameters. Steers fed HTDN diet increased (p = 0.015) the proportion of stearic acid and tended to alter linoleic acid. Overall, saturated, unsaturated, monounsaturated, and polyunsaturated fatty acids of LD muscle were not impacted by the HTDN treatment. A treatment by age interaction was noted for mRNA expression of myosin heavy chain (MHC) IIA, IIX, and stearoyl CoA desaturase (SCD) (p≤0.026). No treatment effect was detected on gene expression from LD muscle biopsies at 7, 18, and 30 mo of age; however, an age effect was detected for all variables measured (p≤0.001). Conclusion Our results indicated that feeding HTDN diet could improve overall quality grade while minimum effects were noted in gene expression, blood parameters, and growing performance. Cattle performance prediction in the feedlot is a critical decision-making tool for optimal planning of cattle fattening and these data provide both benchmark physiological parameters and growth performance measures for Hanwoo cattle feeding enterprises.
Collapse
|
61
|
Explaining Unsaturated Fatty Acids (UFAs), Especially Polyunsaturated Fatty Acid (PUFA) Content in Subcutaneous Fat of Yaks of Different Sex by Differential Proteome Analysis. Genes (Basel) 2022; 13:genes13050790. [PMID: 35627174 PMCID: PMC9140874 DOI: 10.3390/genes13050790] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 01/25/2023] Open
Abstract
Residents on the Tibetan Plateau intake a lot of yak subcutaneous fat by diet. Modern healthy diet ideas demand higher unsaturated fatty acids (UFAs), especially polyunsaturated fatty acid (PUFA) content in meat. Here, the gas chromatography (GC) and tandem mass tag (TMT) proteomic approaches were applied to explore the relationship between the proteomic differences and UFA and PUFA content in the subcutaneous fat of yaks with different sex. Compared with male yaks (MYs), the absolute contents of UFAs, monounsaturated fatty acids (MUFAs) and PUFAs in the subcutaneous fat of female yaks (FYs) were all higher (p < 0.01); the relative content of MUFAs and PUFAs in MY subcutaneous fat was higher, and the value of PUFAs/SFAs was above 0.4, so the MY subcutaneous fat is more healthy for consumers. Further studies showed the transcriptional regulation by peroxisome proliferator-activated receptor delta (PPARD) played a key role in the regulation of UFAs, especially PUFA content in yaks of different sex. In FY subcutaneous fat, the higher abundance of the downstream effector proteins in PPAR signal, including acyl-CoA desaturase (SCD), elongation of very-long-chain fatty acids protein 6 (ELOVL6), lipoprotein lipase (LPL), fatty acid-binding protein (FABP1), very-long-chain (3R)-3-hydroxyacyl-CoA dehydratase 3 (HACD3), long-chain fatty acid CoA ligase 5 (ACSL5) and acyl-CoA-binding protein 2 (ACBP2), promoted the UFAs’ transport and synthesis. The final result was the higher absolute content of c9-C14:1, c9-C18:1, c9,c12-C18:2n-6, c9, c12, c15-C18:3n-3, c5, c8, c11, c14, c17-C20:5n-3, c4, c7, c10, c13, -c16, c19-C22:6n-3, UFAs, MUFAs and PUFAs in FY subcutaneous fat. Further, LPL, FABP1, HACD3, ACSL1 and ACBP2 were the potential biomarkers for PUFA contents in yak subcutaneous fat. This study provides new insights into the molecular mechanisms associated with UFA contents in yak subcutaneous fat.
Collapse
|
62
|
Stearoyl-CoA Desaturase inhibition reverses immune, synaptic and cognitive impairments in an Alzheimer's disease mouse model. Nat Commun 2022; 13:2061. [PMID: 35443751 PMCID: PMC9021296 DOI: 10.1038/s41467-022-29506-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
The defining features of Alzheimer’s disease (AD) include alterations in protein aggregation, immunity, lipid metabolism, synapses, and learning and memory. Of these, lipid abnormalities are the least understood. Here, we investigate the role of Stearoyl-CoA desaturase (SCD), a crucial regulator of fatty acid desaturation, in AD pathogenesis. We show that inhibiting brain SCD activity for 1-month in the 3xTg mouse model of AD alters core AD-related transcriptomic pathways in the hippocampus, and that it concomitantly restores essential components of hippocampal function, including dendritic spines and structure, immediate-early gene expression, and learning and memory itself. Moreover, SCD inhibition dampens activation of microglia, key mediators of spine loss during AD and the main immune cells of the brain. These data reveal that brain fatty acid metabolism links AD genes to downstream immune, synaptic, and functional impairments, identifying SCD as a potential target for AD treatment. Alzheimer’s disease (AD) is characterized by lipid abnormalities which are not well understood. Here, the authors investigate the role of Stearoyl-CoA desaturase (SCD) in a mouse model of AD. They show that inhibiting SCD activity induces major brain and immune cell transcriptional changes and restores dendritic structure and learning and memory.
Collapse
|
63
|
Nagata K, Hishikawa D, Sagara H, Saito M, Watanabe S, Shimizu T, Shindou H. Lysophosphatidylcholine acyltransferase 1 controls mitochondrial reactive oxygen species generation and survival of retinal photoreceptor cells. J Biol Chem 2022; 298:101958. [PMID: 35452679 PMCID: PMC9136105 DOI: 10.1016/j.jbc.2022.101958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/31/2022] [Accepted: 04/15/2022] [Indexed: 12/19/2022] Open
Abstract
Due to their high energy demands and characteristic morphology, retinal photoreceptor cells require a specialized lipid metabolism for survival and function. Accordingly, dysregulation of lipid metabolism leads to the photoreceptor cell death and retinal degeneration. Mice bearing a frameshift mutation in the gene encoding lysophosphatidylcholine acyltransferase 1 (Lpcat1), which produces saturated phosphatidylcholine (PC) composed of two saturated fatty acids, has been reported to cause spontaneous retinal degeneration in mice; however, the mechanism by which this mutation affects degeneration is unclear. In this study, we performed a detailed characterization of LPCAT1 in the retina and found that genetic deletion of Lpcat1 induces light-independent and photoreceptor-specific apoptosis in mice. Lipidomic analyses of the retina and isolated photoreceptor outer segment (OS) suggested that loss of Lpcat1 not only decreased saturated PC production but also affected membrane lipid composition, presumably by altering saturated fatty acyl-CoA availability. Furthermore, we demonstrated that Lpcat1 deletion led to increased mitochondrial reactive oxygen species levels in photoreceptor cells, but not in other retinal cells, and did not affect the OS structure or trafficking of OS-localized proteins. These results suggest that the LPCAT1-dependent production of saturated PC plays critical roles in photoreceptor maturation. Our findings highlight the therapeutic potential of saturated fatty acid metabolism in photoreceptor cell degeneration-related retinal diseases.
Collapse
Affiliation(s)
- Katsuyuki Nagata
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Daisuke Hishikawa
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hiroshi Sagara
- Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Masamichi Saito
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| | - Takao Shimizu
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan; Department of Lipid Science, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
64
|
LncRNA MEG3 up-regulates SIRT6 by ubiquitinating EZH2 and alleviates nonalcoholic fatty liver disease. Cell Death Dis 2022; 8:103. [PMID: 35256601 PMCID: PMC8901640 DOI: 10.1038/s41420-022-00889-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/13/2022] [Accepted: 02/10/2022] [Indexed: 12/24/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a global health threat. Here, we presented the significant role of a novel signaling axis comprising long non-coding RNA maternally expressed gene 3 (MEG3), enhancer of zeste homolog 2 (EZH2), and sirtuin 6 (SIRT6) in controlling lipid accumulation, inflammation, and the progression of NAFLD. Mice fed with high-fat diet (HFD) were established as in vitro and in vivo NAFLD models, respectively. Lipid accumulation was measured by oil red O staining and assays for triglycerides or cholesterol. Inflammation was examined by ELISA for pro-inflammatory cytokines. Gene expressions were examined by RT-qPCR or Western blot. Interactions between key signaling molecules were examined by combining expressional analysis, RNA immunoprecipitation, cycloheximide stability assay, co-immunoprecipitation, and chromatin immunoprecipitation. MEG3 level was reduced in FFA-challenged hepatocytes or liver from HFD-fed mice, and the reduction paralleled the severity of NAFLD in clinic. Overexpressing MEG3 suppressed FFA-induced lipid accumulation or inflammation in hepatocytes. By promoting the ubiquitination and degradation of EZH2, MEG3 upregulated SIRT6, an EZH2 target. SIRT6 essentially mediated the protective effects of MEG3 in hepatocytes. Consistently, overexpressing MEG3 alleviated HFD-induced NAFLD in vivo. By controlling the expressions of genes involved in lipid metabolism and inflammation, the MEG3/EZH2/SIRT6 axis significantly suppressed lipid accumulation and inflammation in vitro, and NAFLD development in vivo. Therefore, boosting MEG3 level may benefit the treatment of NAFLD.
Collapse
|
65
|
Puengel T, Liu H, Guillot A, Heymann F, Tacke F, Peiseler M. Nuclear Receptors Linking Metabolism, Inflammation, and Fibrosis in Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2022; 23:ijms23052668. [PMID: 35269812 PMCID: PMC8910763 DOI: 10.3390/ijms23052668] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) and its progressive form nonalcoholic steatohepatitis (NASH) comprise a spectrum of chronic liver diseases in the global population that can lead to end-stage liver disease and hepatocellular carcinoma (HCC). NAFLD is closely linked to the metabolic syndrome, and comorbidities such as type 2 diabetes, obesity and insulin resistance aggravate liver disease, while NAFLD promotes cardiovascular risk in affected patients. The pathomechanisms of NAFLD are multifaceted, combining hepatic factors including lipotoxicity, mechanisms of cell death and liver inflammation with extrahepatic factors including metabolic disturbance and dysbiosis. Nuclear receptors (NRs) are a family of ligand-controlled transcription factors that regulate glucose, fat and cholesterol homeostasis and modulate innate immune cell functions, including liver macrophages. In parallel with metabolic derangement in NAFLD, altered NR signaling is frequently observed and might be involved in the pathogenesis. Therapeutically, clinical data indicate that single drug targets thus far have been insufficient for reaching patient-relevant endpoints. Therefore, combinatorial treatment strategies with multiple drug targets or drugs with multiple mechanisms of actions could possibly bring advantages, by providing a more holistic therapeutic approach. In this context, peroxisome proliferator-activated receptors (PPARs) and other NRs are of great interest as they are involved in wide-ranging and multi-organ activities associated with NASH progression or regression. In this review, we summarize recent advances in understanding the pathogenesis of NAFLD, focusing on mechanisms of cell death, immunometabolism and the role of NRs. We outline novel therapeutic strategies and discuss remaining challenges.
Collapse
Affiliation(s)
- Tobias Puengel
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
| | - Hanyang Liu
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
| | - Felix Heymann
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
- Correspondence: (F.T.); (M.P.)
| | - Moritz Peiseler
- Department of Hepatology & Gastroenterology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum and Campus Charité Mitte, 13353 Berlin, Germany; (T.P.); (H.L.); (A.G.); (F.H.)
- Berlin Institute of Health (BIH), 10178 Berlin, Germany
- Correspondence: (F.T.); (M.P.)
| |
Collapse
|
66
|
Wang SY, Hu QC, Wu T, Xia J, Tao XA, Cheng B. Abnormal lipid synthesis as a therapeutic target for cancer stem cells. World J Stem Cells 2022; 14:146-162. [PMID: 35432735 PMCID: PMC8963380 DOI: 10.4252/wjsc.v14.i2.146] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/19/2021] [Accepted: 02/20/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer stem cells (CSCs) comprise a subpopulation of cancer cells with stem cell properties, which exhibit the characteristics of high tumorigenicity, self-renewal, and tumor initiation and are associated with the occurrence, metastasis, therapy resistance, and relapse of cancer. Compared with differentiated cells, CSCs have unique metabolic characteristics, and metabolic reprogramming contributes to the self-renewal and maintenance of stem cells. It has been reported that CSCs are highly dependent on lipid metabolism to maintain stemness and satisfy the requirements of biosynthesis and energy metabolism. In this review, we demonstrate that lipid anabolism alterations promote the survival of CSCs, including de novo lipogenesis, lipid desaturation, and cholesterol synthesis. In addition, we also emphasize the molecular mechanism underlying the relationship between lipid synthesis and stem cell survival, the signal trans-duction pathways involved, and the application prospect of lipid synthesis reprogramming in CSC therapy. It is demonstrated that the dependence on lipid synthesis makes targeting of lipid synthesis metabolism a promising therapeutic strategy for eliminating CSCs. Targeting key molecules in lipid synthesis will play an important role in anti-CSC therapy.
Collapse
Affiliation(s)
- Si-Yu Wang
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Qin-Chao Hu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Tong Wu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Juan Xia
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Xiao-An Tao
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| | - Bin Cheng
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou 510000, Guangdong Province, China
| |
Collapse
|
67
|
Landowski M, Bhute VJ, Takimoto T, Grindel S, Shahi PK, Pattnaik BR, Ikeda S, Ikeda A. A mutation in transmembrane protein 135 impairs lipid metabolism in mouse eyecups. Sci Rep 2022; 12:756. [PMID: 35031662 PMCID: PMC8760256 DOI: 10.1038/s41598-021-04644-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/28/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is a significant factor in the development of age-related diseases but how aging disrupts cellular homeostasis to cause age-related retinal disease is unknown. Here, we further our studies on transmembrane protein 135 (Tmem135), a gene involved in retinal aging, by examining the transcriptomic profiles of wild-type, heterozygous and homozygous Tmem135 mutant posterior eyecup samples through RNA sequencing (RNA-Seq). We found significant gene expression changes in both heterozygous and homozygous Tmem135 mutant mouse eyecups that correlate with visual function deficits. Further analysis revealed that expression of many genes involved in lipid metabolism are changed due to the Tmem135 mutation. Consistent with these changes, we found increased lipid accumulation in mutant Tmem135 eyecup samples. Since mutant Tmem135 mice have similar ocular pathologies as human age-related macular degeneration (AMD) eyes, we compared our homozygous Tmem135 mutant eyecup RNA-Seq dataset with transcriptomic datasets of human AMD donor eyes. We found similar changes in genes involved in lipid metabolism between the homozygous Tmem135 mutant eyecups and AMD donor eyes. Our study suggests that the Tmem135 mutation affects lipid metabolism as similarly observed in human AMD eyes, thus Tmem135 mutant mice can serve as a good model for the role of dysregulated lipid metabolism in AMD.
Collapse
Affiliation(s)
- Michael Landowski
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Vijesh J Bhute
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemical Engineering, Imperial College London, South Kensington, London, SW7 2AZ, UK
| | - Tetsuya Takimoto
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Samuel Grindel
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Pawan K Shahi
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Bikash R Pattnaik
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Sakae Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA.
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
68
|
Mengeste AM, Lund J, Katare P, Ghobadi R, Bakke HG, Lunde PK, Eide L, Mahony GO, Göpel S, Peng XR, Kase ET, Thoresen GH, Rustan AC. The small molecule SERCA activator CDN1163 increases energy metabolism in human skeletal muscle cells. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100060. [PMID: 34909682 PMCID: PMC8663964 DOI: 10.1016/j.crphar.2021.100060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 09/20/2021] [Indexed: 12/14/2022] Open
Abstract
Background and objective A number of studies have highlighted muscle-specific mechanisms of thermogenesis involving futile cycling of Ca2+ driven by sarco (endo)plasmic reticulum Ca2+-ATPase (SERCA) and generating heat from ATP hydrolysis to be a promising strategy to counteract obesity and metabolic dysfunction. However, to the best of our knowledge, no experimental studies concerning the metabolic effects of pharmacologically targeting SERCA in human skeletal muscle cells have been reported. Thus, in the present study, we aimed to explore the effects of SERCA-activating compound, CDN1163, on energy metabolism in differentiated human skeletal muscle cells (myotubes). Methods In this study, we used primary myotube cultures derived from muscle biopsies of the musculus vastus lateralis and musculi interspinales from lean, healthy male donors. Energy metabolism in myotubes was studied using radioactive substrates. Oxygen consumption rate was assessed with the Seahorse XF24 bioanalyzer, whereas metabolic genes and protein expressions were determined by qPCR and immunoblotting, respectively. Results Both acute (4 h) and chronic (5 days) treatment of myotubes with CDN1163 showed increased uptake and oxidation of glucose, as well as complete fatty acid oxidation in the presence of carbonyl cyanide 4-(trifluromethoxy)phenylhydrazone (FCCP). These effects were supported by measurement of oxygen consumption rate, in which the oxidative spare capacity and maximal respiration were enhanced after CDN1163-treatment. In addition, chronic treatment with CDN1163 improved cellular uptake of oleic acid (OA) and fatty acid β-oxidation. The increased OA metabolism was accompanied by enhanced mRNA-expression of carnitine palmitoyl transferase (CPT) 1B, pyruvate dehydrogenase kinase (PDK) 4, as well as increased AMP-activated protein kinase (AMPK)Thr172 phosphorylation. Moreover, following chronic CDN1163 treatment, the expression levels of stearoyl-CoA desaturase (SCD) 1 was decreased together with de novo lipogenesis from acetic acid and formation of diacylglycerol (DAG) from OA. Conclusion Altogether, these results suggest that SERCA activation by CDN1163 enhances energy metabolism in human myotubes, which might be favourable in relation to disorders that are related to metabolic dysfunction such as obesity and type 2 diabetes mellitus. CDN1163 induced an increase in glucose and fatty acid metabolism in primary human myotubes. Myotubes treated with CDN1163 showed lower intramyocellular lipid accumulation and higher rate of β-oxidation. AMPK activity was upregulated in CDN1163-treated myotubes.
Collapse
Key Words
- AMPK
- AMPK, AMP-activated protein kinase
- ASM, acid-soluble metabolites
- CE, cholesteryl ester
- DAG, diacylglycerol
- FA, fatty acid
- FCCP, 4-(trifluromethoxy)phenylhydrazone
- Glucose metabolism
- Lipid metabolism
- OA, oleic acid
- OCR, oxygen consumption rate
- Obesity
- SCD1, stearoyl-CoA desaturase 1
- SERCA
- SERCA, sarco(endo)plasmic reticulum Ca2+-ATPase
- Skeletal muscle
- T2DM, type 2 diabetes mellitus
- Type 2 diabetes
Collapse
Affiliation(s)
- Abel M Mengeste
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Jenny Lund
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Parmeshwar Katare
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Roya Ghobadi
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Hege G Bakke
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - Per Kristian Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Norway.,KG Jebsen Cardiac Research Centre, University of Oslo, Norway
| | - Lars Eide
- Department of Medical Biochemistry, Institute of Clinical Medicine, University of Oslo, Norway
| | - Gavin O' Mahony
- Medicinal Chemistry, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Sven Göpel
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Xiao-Rong Peng
- Bioscience Metabolism, Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Eili Tranheim Kase
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| | - G Hege Thoresen
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway.,Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Norway
| | - Arild C Rustan
- Section for Pharmacology and Pharmaceutical Biosciences, Department of Pharmacy, University of Oslo, Norway
| |
Collapse
|
69
|
Roškarić P, Šperanda M, Mašek T, Verbanac D, Starčević K. Low Dietary n6/n3 Ratio Attenuates Changes in the NRF 2 Gene Expression, Lipid Peroxidation, and Inflammatory Markers Induced by Fructose Overconsumption in the Rat Abdominal Adipose Tissue. Antioxidants (Basel) 2021; 10:2005. [PMID: 34943108 PMCID: PMC8698844 DOI: 10.3390/antiox10122005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022] Open
Abstract
The objective of this study was to examine the benefits of different n6/n3 polyunsaturated fatty acid ratios on the lipid metabolism, insulin resistance, and oxidative stress in the adipose tissue of rats fed a high-fructose diet. Male and female rats were divided into four groups: a control group (CON) (n6/n3 ratio ~7), a high-fructose group (HF) (n6/n3 ratio ~7), an N6-HF group (n6/n3 ratio ~50), and the DHA-HF group (n6/n3 ratio ~1, with the addition of docosahexaenoic (DHA) and eicosapentaenoic (EPA) acid). The CON group received plain water and the HF group received 15% fructose in their drinking water. Fructose induced an increase in the content of serum triglycerides, serum cholesterol, and HOMA-IR index. Among the fatty acids, elevated proportions of C18:1n9 and C16:1n7, as well as an increase in total monounsaturated fatty acid (MUFA), were found in the adipose tissue of the HF group. Fructose treatment also changed oxidative parameters, including a marked increase in the serum malondialdehyde (MDA) content. Meanwhile, DHA supplementation caused a significant decrease in the serum MDA concentration in comparison with the HF group. In addition, DHA/EPA supplementation attenuated oxidative stress by increasing NRF 2 gene expression. Fructose treatment also significantly decreased the adiponectin level, while DHA supplementation ameliorated it. The changes observed in this trial, including the decrease in the content of DHA and EPA, the decreased EPA/ARA ratio, and the increase in the expression of inflammatory genes, are characteristics of the low-grade inflammation caused by fructose treatment. These changes in the rat adipose tissue could be prevented by dietary intervention consisting of DHA supplementation and a low n6/n3 ratio.
Collapse
Affiliation(s)
- Petra Roškarić
- Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia;
| | - Marcela Šperanda
- Department of Animal Science, Faculty of Agriculture, University of Osijek, Vladimira Preloga 1, 31000 Osijek, Croatia;
| | - Tomislav Mašek
- Department of Animal Nutrition and Dietetics, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia;
| | - Donatella Verbanac
- Department of Medical Biochemistry and Hematology, Faculty of Pharmacy and Biochemistry, University of Zagreb, A. Kovačića 1, 10000 Zagreb, Croatia;
| | - Kristina Starčević
- Department of Chemistry and Biochemistry, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10000 Zagreb, Croatia;
| |
Collapse
|
70
|
González-Rovira A, Mourente G, Igartuburu JM, Pendon C. Molecular and functional characterization of a SCD 1b from European sea bass (Dicentrarchus labrax L.). Comp Biochem Physiol B Biochem Mol Biol 2021; 258:110698. [PMID: 34801709 DOI: 10.1016/j.cbpb.2021.110698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 10/19/2022]
Abstract
Fatty acid desaturation is a highly complex and regulated process involving different molecular and genetic actors. Ultimally, the fatty acid desaturase enzymes are responsible for the introduction of double bonds at different positions of specific substrates, resulting in a wide variety of mono- and poly-unsaturated fatty acids. This substrate-specificity makes it possible to meet all the functional needs of the different tissues against a wide variety of internal and external conditions, giving rise to a varied profile of expression and functionality of the different desaturases in the body. Being our main interest to study and characterize at the molecular level the fatty acid desaturation process in fishes, we have focused our effort on characterizing SCD 1b from European sea bass (Dicentrarchus labrax, L.). In this work, we have characterized a tearoyl-CoA Desaturase cDNA that codes a protein of 334 amino acids, which shares the greatest homology to marine fish SCD 1b. Northern blot analysis showed two transcripts of 3.5 kb and 1.4 kb. Two putative cis-acting conserved motifs are localized in the cDNA 5'-end: a polypyrimidine CT dinucleotide repeat tract and two non-palindromic putative NRL-response elements (NREs). The deduced protein presents two Δ9 FADs like domain, three His-rich motifs, a total of nine His residues acting as di‑iron coordination ligands. The SCD 1b 3D protein modelling shows a structure made up primarily of α-helices, four of which could be transmembrane helices. The catalytic region is oriented to the cytosolic side of the Endoplasmic Reticulum membrane, where the 9-histidine residues are arranged coordinated to two non-heme Fe2+ ions. A new His-containing motif NX3H-like includes an Asn residue that participates in the coordination of Fe2+1 through a water molecule. The protein has a large pocket with a large opening to the outside. It includes a tunnel in which the substrate-binding site is located. The external shape is reminiscent of a boathook. It shows group specificity, although a greater preference for 18C substrates. The length of the tunnel, delimited by seven amino acids that forms a pocket at the end of the tunnel, the possibility that the substrates adopt different conformations inside the tunnel as well as and the movement of acyl chain inside the tunnel, could explain the high preference for 18C fatty acids and the group specificity of the enzyme. The cDNA encodes a functional SCD enzyme, whose subcellular localization is the Endoplasmic Reticulum, which complements the ole1Δ gene-disrupted gene in DTY-11A Saccharomyces cerevisiae strain and produces an increment of palmitoleic and oleic acids. The scd 1b gene is expressed in all tested tissues, showing the liver and adipose tissue a higher level of expression against the brain, heart, gonad and intestine. Scd 1b expression was always bigger than those of the Δ6 fad gene, being especially significant in adipose tissue and liver. From our data, we conclude that, in contrast to the functional significance of SCD 1b in adipose tissue, liver and heart, Δ6 FAD seems to play a more determining role in the biosynthesis of unsaturated fatty acids in the intestine, brain and gonad in fish.
Collapse
Affiliation(s)
- Almudena González-Rovira
- Departamento de Biomedicina, Biotecnología y Salud Pública, INBIO, Facultad de Ciencias, Universidad de Cádiz, 11519 Puerto Real, Cádiz, Spain.
| | - Gabriel Mourente
- Departamento de Biología, Facultad de Ciencias del Mar y Ambientales, Universidad de Cádiz, 11519 Puerto Real, Cádiz, Spain.
| | - José Manuel Igartuburu
- Departamento de Química Orgánica, INBIO, Facultad de Ciencias, Universidad de Cádiz, 11519 Puerto Real, Cádiz, Spain.
| | - Carlos Pendon
- Departamento de Biomedicina, Biotecnología y Salud Pública, INBIO, Facultad de Ciencias, Universidad de Cádiz, 11519 Puerto Real, Cádiz, Spain.
| |
Collapse
|
71
|
Demerdash HM. Weight regain after bariatric surgery: Promoters and potential predictors. World J Meta-Anal 2021; 9:438-454. [DOI: 10.13105/wjma.v9.i5.438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/07/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
Obesity is globally viewed as chronic relapsing disease. Bariatric surgery offers the most efficient and durable weight loss approach. However, weight regain after surgery is a distressing issue as obesity can revert. Surgical procedures were originally designed to reduce food intake and catalyze weight loss, provided that its role is marginalized in long-term weight maintenance. Consequently, it is essential to establish a scientifically standardized applicable definitions for weight regain, which necessitates enhanced comprehension of the clinical situation, as well as have realistic expectations concerning weight loss. Moreover, several factors are proposed to influence weight regain as psychological, behavioral factors, hormonal, metabolic, anatomical lapses, as well as genetic predisposition. Recently, there is a growing evidence of utilization of scoring system to anticipate excess body weight loss, along with characterizing certain biomarkers that identify subjects at risk of suboptimal weight loss after surgery. Furthermore, personalized counseling is warranted to help select bariatric procedure, reinforce self-monitoring skills, motivate patient, encourage mindful eating practices, to avoid recidivism.
Collapse
Affiliation(s)
- Hala Mourad Demerdash
- Department of Clinical Pathology, Alexandria University Hospitals, Alexandria 21311, Egypt
| |
Collapse
|
72
|
Raman microscopy-based quantification of the physical properties of intracellular lipids. Commun Biol 2021; 4:1176. [PMID: 34625633 PMCID: PMC8501034 DOI: 10.1038/s42003-021-02679-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023] Open
Abstract
The physical properties of lipids, such as viscosity, are homeostatically maintained in cells and are intimately involved in physiological roles. Measurement of the physical properties of plasma membranes has been achieved primarily through chemical or genetically encoded fluorescent probes. However, since most probes target plasma membranes, physical properties of lipids in intracellular organelles, including lipid droplets (LDs) are yet to be analyzed. Here, we present a novel Raman microscopy-based approach for quantifying the physical properties of intracellular lipids under deuterium-labeled fatty acid treatment conditions. Focusing on the fact that Raman spectra of carbon-deuterium vibration are altered depending on the surrounding lipid species, we quantitatively represented the physical properties of lipids as the gauche/trans conformational ratio of the introduced labeled fatty acids, which can be used as an indicator of viscosity. Intracellular Raman imaging revealed that the gauche/trans ratio of cytosolic regions was robustly preserved against perturbations attempting to alter the lipid composition. This was likely due to LDs functioning as a buffer against excess gauche/trans ratio, beyond its traditional role as an energy storage organelle. Our novel approach enables the observation of the physical properties of organelle lipids, which is difficult to perform with conventional probes, and is useful for quantitative assessment of the subcellular lipid environment.
Collapse
|
73
|
Bai J, Li J, Pan R, Zhu Y, Xiao X, Li Y, Li C. Polysaccharides from Volvariella volvacea inhibit fat accumulation in C. elegans dependent on the aak-2/nhr-49-mediated pathway. J Food Biochem 2021; 45:e13912. [PMID: 34561881 DOI: 10.1111/jfbc.13912] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 08/02/2021] [Accepted: 08/09/2021] [Indexed: 12/28/2022]
Abstract
Volvariella volvacea has bioactivities in improving immunity, anti-oxidation, and alleviating obesity, which is an excellent functional food. Polysaccharide from Volvariella volvacea (VPS), one of the main bioactive components, exerts a potential fat-lowering effect, but its exact mechanism remains unclear. In this study, the effects and molecular pathways of VPS regulate the fat deposition of Caenorhabditis elegans. Results showed that VPS at low (250 μg/ml), medium (500 μg/ml) and high (750 μg/ml) concentrations all reduced the overall fat, without inhibitory effects on the growth and movement abilities of nematode. VPS at 500 μg/ml could dramatically decrease the triglyceride (TG) level of wild-type nematode, while no significant changes in TG content were observed in mutants deficient in aak-2 (energy receptor), nhr-49 (nuclear transcription factor), fat-5, and fat-7 genes. VPS declines fat storage of C. elegans, largely through the aak-2/nhr-49-mediated fatty acid synthesis pathway, and partially the acs-2-mediated fatty acid oxidation pathway. PRACTICAL APPLICATIONS: A model illustrates the mechanism of polysaccharide from Volvariella volvacea (VPS) inhibiting fat accumulation in Caenorhabditis elegans. VPS may directly or indirectly activate the energy sensor aak-2, which governs lipid metabolism. Results demonstrate that VPS regulates fat metabolism including fatty acid oxidation (FAO) and fatty acid synthesis (FAS), rather than lipolysis. In the FAO, VPS promotes FAO by up-regulating the mRNA and protein levels of acs-2. In FAS, VPS significantly down-regulated the transcriptional regulator nhr-49 and the downstream targets fat-5, fat-6, and fat-7, thereby declining the overall fat deposition. In conclusion, VPS inhibits the fat accumulation of C. elegans largely dependent on an aak-2/nhr-49-mediated FAS pathway.
Collapse
Affiliation(s)
- Juan Bai
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun, China.,School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China.,Jiangsu Jiangnan Biotechnology Co., Ltd., Zhenjiang, China
| | - Jie Li
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Ruirong Pan
- Department of Endocrinology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Ying Zhu
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Xiang Xiao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yu Li
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun, China
| | - Changtian Li
- Engineering Research Center of Edible and Medicinal Fungi, Ministry of Education, Jilin Agricultural University, Changchun, China
| |
Collapse
|
74
|
Clifford BL, Sedgeman LR, Williams KJ, Morand P, Cheng A, Jarrett KE, Chan AP, Brearley-Sholto MC, Wahlström A, Ashby JW, Barshop W, Wohlschlegel J, Calkin AC, Liu Y, Thorell A, Meikle PJ, Drew BG, Mack JJ, Marschall HU, Tarling EJ, Edwards PA, de Aguiar Vallim TQ. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab 2021; 33:1671-1684.e4. [PMID: 34270928 PMCID: PMC8353952 DOI: 10.1016/j.cmet.2021.06.012] [Citation(s) in RCA: 246] [Impact Index Per Article: 61.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/12/2021] [Accepted: 06/21/2021] [Indexed: 12/24/2022]
Abstract
FXR agonists are used to treat non-alcoholic fatty liver disease (NAFLD), in part because they reduce hepatic lipids. Here, we show that FXR activation with the FXR agonist GSK2324 controls hepatic lipids via reduced absorption and selective decreases in fatty acid synthesis. Using comprehensive lipidomic analyses, we show that FXR activation in mice or humans specifically reduces hepatic levels of mono- and polyunsaturated fatty acids (MUFA and PUFA). Decreases in MUFA are due to FXR-dependent repression of Scd1, Dgat2, and Lpin1 expression, which is independent of SHP and SREBP1c. FXR-dependent decreases in PUFAs are mediated by decreases in lipid absorption. Replenishing bile acids in the diet prevented decreased lipid absorption in GSK2324-treated mice, suggesting that FXR reduces absorption via decreased bile acids. We used tissue-specific FXR KO mice to show that hepatic FXR controls lipogenic genes, whereas intestinal FXR controls lipid absorption. Together, our studies establish two distinct pathways by which FXR regulates hepatic lipids.
Collapse
Affiliation(s)
- Bethan L Clifford
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Leslie R Sedgeman
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Kevin J Williams
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Lipidomics Core Facility, Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Pauline Morand
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Angela Cheng
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Kelsey E Jarrett
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Alvin P Chan
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Madelaine C Brearley-Sholto
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Annika Wahlström
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Julianne W Ashby
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - William Barshop
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - James Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Anna C Calkin
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia; Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Yingying Liu
- Lipid Metabolism & Cardiometabolic Disease Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia; Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Anders Thorell
- Karolinska Institutet, Department of Clinical Science, Danderyd Hospital and Department of Surgery, Ersta Hospital, Stockholm, Sweden
| | - Peter J Meikle
- Metabolomics Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Brian G Drew
- Central Clinical School, Monash University, Melbourne, VIC, Australia; Molecular Metabolism & Ageing Laboratory, Baker Heart & Diabetes Institute, Melbourne, VIC, Australia
| | - Julia J Mack
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Hanns-Ulrich Marschall
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elizabeth J Tarling
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center (JCCC), UCLA, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Peter A Edwards
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA
| | - Thomas Q de Aguiar Vallim
- Department of Medicine, Division of Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center (JCCC), UCLA, Los Angeles, CA, USA; Molecular Biology Institute, University of California, Los Angeles (UCLA), Los Angeles, CA, USA.
| |
Collapse
|
75
|
Akinci B, Subauste A, Ajluni N, Esfandiari NH, Meral R, Neidert AH, Eraslan A, Hench R, Rus D, Mckenna B, Hussain HK, Chenevert TL, Tayeh MK, Rupani AR, Innis JW, Mantzoros CS, Conjeevaram HS, Burant CL, Oral EA. Metreleptin therapy for nonalcoholic steatohepatitis: Open-label therapy interventions in two different clinical settings. MED 2021; 2:814-835. [DOI: 10.1016/j.medj.2021.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
76
|
Yuan X, Hu S, Li L, Han C, Liu H, He H, Xia L, Hu J, Hu B, Ran M, Liu Y, Wang J. Lipidomics profiling of goose granulosa cell model of stearoyl-CoA desaturase function identifies a pattern of lipid droplets associated with follicle development. Cell Biosci 2021; 11:95. [PMID: 34022953 PMCID: PMC8141238 DOI: 10.1186/s13578-021-00604-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/04/2021] [Indexed: 11/23/2022] Open
Abstract
Background Despite their important functions and nearly ubiquitous presence in cells, an understanding of the biology of intracellular lipid droplets (LDs) in goose follicle development remains limited. An integrated study of lipidomic and transcriptomic analyses was performed in a cellular model of stearoyl-CoA desaturase (SCD) function, to determine the effects of intracellular LDs on follicle development in geese. Results Numerous internalized LDs, which were generally spherical in shape, were dispersed throughout the cytoplasm of granulosa cells (GCs), as determined using confocal microscopy analysis, with altered SCD expression affecting LD content. GC lipidomic profiling showed that the majority of the differentially abundant lipid classes were glycerophospholipids, including PA, PC, PE, PG, PI, and PS, and glycerolipids, including DG and TG, which enriched glycerophospholipid, sphingolipid, and glycerolipid metabolisms. Furthermore, transcriptomics identified differentially expressed genes (DEGs), some of which were assigned to lipid-related Gene Ontology slim terms. More DEGs were assigned in the SCD-knockdown group than in the SCD-overexpression group. Integration of the significant differentially expressed genes and lipids based on pathway enrichment analysis identified potentially targetable pathways related to glycerolipid/glycerophospholipid metabolism. Conclusions This study demonstrated the importance of lipids in understanding follicle development, thus providing a potential foundation to decipher the underlying mechanisms of lipid-mediated follicle development. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00604-6.
Collapse
Affiliation(s)
- Xin Yuan
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Shenqiang Hu
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Liang Li
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Chunchun Han
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Hehe Liu
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Hua He
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Lu Xia
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Jiwei Hu
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Bo Hu
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Mingxia Ran
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Yali Liu
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Jiwen Wang
- Country Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China.
| |
Collapse
|
77
|
Igarashi Y, Iida S, Dai J, Huo J, Cui X, Sawashita J, Mori M, Miyahara H, Higuchi K. Glavonoid-rich oil supplementation reduces stearoyl-coenzyme A desaturase 1 expression and improves systemic metabolism in diabetic, obese KK-A y mice. Biomed Pharmacother 2021; 140:111714. [PMID: 34022607 DOI: 10.1016/j.biopha.2021.111714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
AIMS Glavonoid-rich oil (GRO) derived from ethanol extraction of licorice (Glycyrrhiza glabra Linne) root has been reported to have beneficial effects on health. In this study, we aimed to determine the effect of long-term administration of GRO on metabolic disorders and to elucidate the molecular mechanism. MAIN METHODS Female obese, type 2 diabetic KK-Ay mice were fed diets supplemented with 0.3% or 0.8% GRO (w/w) for 4-12 weeks. Mice were euthanized and autopsied at 20 weeks old. The effects of GRO on lipid and glucose metabolism were evaluated by measuring physiological and biochemical markers using mRNA sequencing, quantitative reverse-transcription PCR, and western blot analyses. KEY FINDINGS Compared to mice fed the control diet, GRO-supplemented mice had reduced body and white adipose tissue weights, serum levels of triglycerides and cholesterol, and improved glucose tolerance, while food intake was not affected. We found remarkable reductions in the gene expression levels of stearoyl-coenzyme A desaturase 1 (Scd1) and pyruvate dehydrogenase kinase isoenzyme 4 (Pdk4) in the liver, in addition to decreased expression of fatty acid synthase (Fasn) in inguinal white adipose tissue (iWAT). These results suggest that GRO supplementation improves lipid profiles via reduced de novo lipogenesis in the liver and white adipose tissue. Glucose metabolism may also be improved by increased glycolysis in the liver. SIGNIFICANCE Our analysis of long-term supplementation of GRO in obese and diabetic mice should provide novel insight into preventing insulin resistance and metabolic syndromes.
Collapse
Affiliation(s)
- Yuichi Igarashi
- Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Shiho Iida
- Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Jian Dai
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Jia Huo
- Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan; The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoran Cui
- Department of Aging Biology, Shinshu University Graduate School of Medicine, Matsumoto, Japan
| | - Jinko Sawashita
- Research & Development Team, Supplement Business Division, Pharma & Supplemental Nutrition Solutions Vehicle, Kaneka Corporation, Osaka, Japan
| | - Masayuki Mori
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan; Department of Aging Biology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Hiroki Miyahara
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan
| | - Keiichi Higuchi
- Department of Neuro-Health Innovation, Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, Matsumoto, Japan; Department of Aging Biology, Shinshu University School of Medicine, Matsumoto, Japan; Community Health Care Research Center, Nagano University of Health and Medicine, Nagano, Japan.
| |
Collapse
|
78
|
Extraction and Hypolipidemic Activity of Low Molecular Weight Polysaccharides Isolated from Rosa Laevigata Fruits. BIOMED RESEARCH INTERNATIONAL 2021; 2020:2043785. [PMID: 33145340 PMCID: PMC7599405 DOI: 10.1155/2020/2043785] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 12/25/2022]
Abstract
Three novel low molecular weight polysaccharides (RLP-1a, RLP-2a, and RLP-3a) with 9004, 8761, and 7571 Da were first obtained by purifying the crude polysaccharides from the fruits of a traditional Chinese medicinal herb Rosae Laevigatae. The conditions for polysaccharides from the R. Laevigatae fruit (RLP) extraction were optimized by the response surface methodology, and the optimal conditions were as follows: extraction temperature, 93°C; extraction time, 2.8 h; water to raw material ratio, 22; extraction frequency, 3. Structural characterization showed that RLP-1a consisted of rhamnose, arabinose, xylose, glucose, and galactose with the ratio of 3.14 : 8.21 : 1 : 1.37 : 4.90, whereas RLP-2a was composed of rhamnose, mannose, glucose, and galactose with the ratio of 1.70 : 1 : 93.59 : 2.73, and RLP-3a was composed of rhamnose, arabinose, xylose, mannose, glucose, and galactose with the ratio of 6.04 : 26.51 : 2.05 : 1 : 3.17 : 31.77. The NMR analyses revealed that RLP-1a, RLP-2a, and RLP-3a contained 6, 4, and 6 types of glycosidic linkages, respectively. RLP-1a and RLP-3a exhibited distinct antioxidant abilities on the superoxide anions, 1,1-diphenyl-2-picrylhydrazyl (DPPH), and hydroxyl radicals in vitro. RLPs could decrease the serum lipid levels, elevate the serum high-density lipoprotein cholesterol levels, enhance the antioxidant enzymes levels, and upregulate of FADS2, ACOX3, and SCD-1 which involved in the lipid metabolic processes and oxidative stress in the high-fat diet-induced rats. These results suggested that RLPs ameliorated the high-fat diet- (HFD-) induced lipid metabolism disturbance in the rat liver through the peroxisome proliferator-activated receptor (PPAR) signaling pathway. Low molecular weight polysaccharides of RLP could be served as a novel potential functional food for improving hyperlipidemia and liver oxidative stress responses.
Collapse
|
79
|
Peng Y, Li H, Liu Z, Zhang C, Li K, Gong Y, Geng L, Su J, Guan X, Liu L, Zhou R, Zhao Z, Guo J, Liang Q, Li X. Chromosome-level genome assembly of the Arctic fox (Vulpes lagopus) using PacBio sequencing and Hi-C technology. Mol Ecol Resour 2021; 21:2093-2108. [PMID: 33829635 DOI: 10.1111/1755-0998.13397] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 03/28/2021] [Accepted: 03/30/2021] [Indexed: 10/21/2022]
Abstract
The Arctic fox (Vulpes lagopus) is the only fox species occurring in the Arctic and has adapted to its extreme climatic conditions. Currently, the molecular basis of its adaptation to the extreme climate has not been characterized. Here, we applied PacBio sequencing and chromosome structure capture technique to assemble the first V. lagopus genome assembly, which is assembled into chromosome fragments. The genome assembly has a total length of 2.345 Gb with a contig N50 of 31.848 Mb and a scaffold N50 of 131.537 Mb, consisting of 25 pseudochromosomal scaffolds. The V. lagopus genome had approximately 32.33% repeat sequences. In total, 21,278 protein-coding genes were predicted, of which 99.14% were functionally annotated. Compared with 12 other mammals, V. lagopus was most closely related to V. Vulpes with an estimated divergence time of ~7.1 Ma. The expanded gene families and positively selected genes potentially play roles in the adaptation of V. lagopus to Arctic extreme environment. This high-quality assembled genome will not only promote future studies of genetic diversity and evolution in foxes and other canids but also provide important resources for conservation of Arctic species.
Collapse
Affiliation(s)
- Yongdong Peng
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Hong Li
- Novogene Bioinformatics Institute, Beijing, China
| | - Zhengzhu Liu
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Chuansheng Zhang
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Keqiang Li
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Mathematics and Information Science, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Yuanfang Gong
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Liying Geng
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Agronomy and Biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Jingjing Su
- Hebei Key Laboratory of Applied Chemistry, School of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao, China
| | - Xuemin Guan
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Agronomy and Biotechnology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Lei Liu
- College of Animal Science and Technology, Shandong Agricultural University, Tai-an, China
| | - Ruihong Zhou
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Ziya Zhao
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Jianxu Guo
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| | - Qiqi Liang
- Novogene Bioinformatics Institute, Beijing, China
| | - Xianglong Li
- Hebei Key Laboratory of Specialty Animal Germplasm Resources Exploration and Innovation (Under Planning), College of Animal Science and Technology, Hebei Normal University of Science and Technology, Qinhuangdao, China
| |
Collapse
|
80
|
Pinterić M, Podgorski II, Popović Hadžija M, Tartaro Bujak I, Tadijan A, Balog T, Sobočanec S. Chronic High Fat Diet Intake Impairs Hepatic Metabolic Parameters in Ovariectomized Sirt3 KO Mice. Int J Mol Sci 2021; 22:ijms22084277. [PMID: 33924115 PMCID: PMC8074326 DOI: 10.3390/ijms22084277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/14/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
High fat diet (HFD) is an important factor in the development of metabolic diseases, with liver as metabolic center being highly exposed to its influence. However, the effect of HFD-induced metabolic stress with respect to ovary hormone depletion and sirtuin 3 (Sirt3) is not clear. Here we investigated the effect of Sirt3 in liver of ovariectomized and sham female mice upon 10 weeks of feeding with standard-fat diet (SFD) or HFD. Liver was examined by Folch, gas chromatography and lipid hydroperoxide analysis, histology and oil red staining, RT-PCR, Western blot, antioxidative enzyme and oxygen consumption analyses. In SFD-fed WT mice, ovariectomy increased Sirt3 and fatty acids synthesis, maintained mitochondrial function, and decreased levels of lipid hydroperoxides. Combination of ovariectomy and Sirt3 depletion reduced pparα, Scd-1 ratio, MUFA proportions, CII-driven respiration, and increased lipid damage. HFD compromised CII-driven respiration and activated peroxisomal ROS scavenging enzyme catalase in sham mice, whereas in combination with ovariectomy and Sirt3 depletion, increased body weight gain, expression of NAFLD- and oxidative stress-inducing genes, and impaired response of antioxidative system. Overall, this study provides evidence that protection against harmful effects of HFD in female mice is attributed to the combined effect of female sex hormones and Sirt3, thus contributing to preclinical research on possible sex-related therapeutic agents for metabolic syndrome and associated diseases.
Collapse
Affiliation(s)
- Marija Pinterić
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.P.); (I.I.P.); (M.P.H.); (A.T.); (T.B.)
| | - Iva I. Podgorski
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.P.); (I.I.P.); (M.P.H.); (A.T.); (T.B.)
| | - Marijana Popović Hadžija
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.P.); (I.I.P.); (M.P.H.); (A.T.); (T.B.)
| | - Ivana Tartaro Bujak
- Division of Materials Chemistry, Ruđer Bošković Institute, 10000 Zagreb, Croatia;
| | - Ana Tadijan
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.P.); (I.I.P.); (M.P.H.); (A.T.); (T.B.)
| | - Tihomir Balog
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.P.); (I.I.P.); (M.P.H.); (A.T.); (T.B.)
| | - Sandra Sobočanec
- Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia; (M.P.); (I.I.P.); (M.P.H.); (A.T.); (T.B.)
- Correspondence: ; Tel.: +385-1-4561-172
| |
Collapse
|
81
|
Masoudi S, Mitchell TW, Willcox MD. Profiling of non-polar lipids in tears of contact lens wearers during the day. Exp Eye Res 2021; 207:108567. [PMID: 33848523 DOI: 10.1016/j.exer.2021.108567] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE This study explored whether the non-polar lipids in the human tear fluid lipidome show diurnal variation with and without contact lens wear. It also addressed the relationship between changes in ocular comfort during the day with the level of non-polar lipids. METHODS Tear samples were collected in the morning and evening with and without contact lenses using fine glass capillary tubes and were analysed by chip-based nano-electrospray ionization tandem mass spectrometric techniques. Tear levels of cholesteryl esters (CE), wax esters (WE) and triacylglycerides (TAG) were quantified. RESULTS TAG 48:0, 52:0 and WE 26:0/16:0, and 27:0/17:0 increased from morning to evening. TAG 52:2, WE 21:0/16:0, 21:0/18:1 and 28:0/18:1 decreased during the day when no lenses were worn. CE 21:0 was the only non-polar lipid that increased from morning to evening in contact lens wear. WE 21:0/16:0 and 27:0/17:0 were lower in the morning in contact lens wear compared to no lens wear (p ≤ 0.05). The level of non-polar lipids did not correlate with ocular comfort at the end of the day. CONCLUSION Even though the level of some of non-polar lipid species changed from morning to evening the total level of major tear non-polar lipids remained unchanged during the day with and without contact lens wear. The effect of change in the quantity and structure of lipid species on tear stability and ocular comfort warrants more investigation.
Collapse
Affiliation(s)
- Simin Masoudi
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia.
| | - Todd W Mitchell
- School of Medicine and Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia; Illawara Health and Medical Research Institute, Wollongong, New South Wales, Australia.
| | - Mark D Willcox
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia.
| |
Collapse
|
82
|
Kuppusamy P, Ilavenil S, Hwang IH, Kim D, Choi KC. Ferulic Acid Stimulates Adipocyte-Specific Secretory Proteins to Regulate Adipose Homeostasis in 3T3-L1 Adipocytes. Molecules 2021; 26:molecules26071984. [PMID: 33915783 PMCID: PMC8037266 DOI: 10.3390/molecules26071984] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 01/16/2023] Open
Abstract
Obesity has recently emerged as a public health issue facing developing countries in the world. It is caused by the accumulation of fat in adipose, characterized by insulin resistance, excessive lipid accumulation, inflammation, and oxidative stress, leading to an increase in adipokine levels. Herein, we investigated the capacity of a bioactive polyphenolic compound (ferulic acid (FA)) to control adipocyte dysfunction in 3T3-L1 adipocytes (in vitro). Key adipocyte differentiation markers, glycerol content, lipolysis-associated mRNA, and proteins were measured in experimental adipocytes. FA-treated adipocytes exhibited downregulated key adipocyte differentiation factors peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAT enhancer binding-proteins-α (C/EBP-α) and its downstream targets in a time-dependent manner. The FA-treated 3T3-L1 adipocytes showed an increased release of glycerol content compared with non-treated adipocytes. Also, FA treatment significantly up-regulated the lipolysis-related factors, including p-HSL, and p-perilipin, and down-regulated ApoD, Sema3C, Cxcl12, Sfrp2, p-stearoyl-CoA desaturase 1 (SCD1), adiponectin, and Grk5. Also, the FA treatment showed significantly down-regulated adipokines leptin, chemerin, and irisin than the non-treated cells. The present findings indicated that FA showed significant anti-adipogenic and lipogenic activities by regulating key adipocyte factors and enzyme, enhanced lipolysis by HSL/perilipin cascade. FA is considered a potent molecule to prevent obesity and its associated metabolic changes in the future.
Collapse
Affiliation(s)
- Palaniselvam Kuppusamy
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Korea; (P.K.); (S.I.)
| | - Soundharrajan Ilavenil
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Korea; (P.K.); (S.I.)
| | - In Ho Hwang
- Department of Animal Science, College of Agricultural and Life Science, Chonbuk National University, Jeonju 54896, Korea;
| | - Dahye Kim
- Faculty of Biotechnology, College of Applied Life Science, Jeju National University, Jeonju 63294, Korea
- Correspondence: (D.K.); (K.C.C.); Tel.: +82-64-754-3317 (D.K.); +82-41-580-6752 (K.C.C.); Fax: +82-64-756-3348 (D.K.); +82-41-580-6779 (K.C.C.)
| | - Ki Choon Choi
- Grassland and Forage Division, National Institute of Animal Science, Rural Development Administration, Cheonan 330-801, Korea; (P.K.); (S.I.)
- Correspondence: (D.K.); (K.C.C.); Tel.: +82-64-754-3317 (D.K.); +82-41-580-6752 (K.C.C.); Fax: +82-64-756-3348 (D.K.); +82-41-580-6779 (K.C.C.)
| |
Collapse
|
83
|
Effects of Ethanol Feeding in Early-Stage NAFLD Mice Induced by Western Diet. LIVERS 2021. [DOI: 10.3390/livers1010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: The prevalence of metabolic liver diseases is increasing and approved pharmacological treatments are still missing. Many animal models of nonalcoholic fatty liver disease (NAFLD) show a full spectrum of fibrosis, inflammation and steatosis, which does not reflect the human situation since only up to one third of the patients develop fibrosis and nonalcoholic steatohepatitis (NASH). Methods: Seven week old C57Bl/J mice were treated with ethanol, Western diet (WD) or both. The animals’ liver phenotypes were determined through histology, immunohistochemistry, Western blotting, hepatic triglyceride content and gene expression levels. In a human cohort of 80 patients stratified by current alcohol misuse and body mass index, liver histology and gene expression analysis were performed. Results: WD diet and ethanol-treated animals showed severe steatosis, with high hepatic triglyceride content and upregulation of fatty acid synthesis. Mild fibrosis was revealed using Sirius-red stains and gene expression levels of collagen. Inflammation was detected using histology, immunohistochemistry and upregulation of proinflammatory genes. The human cohort of obese drinkers showed similar upregulation in genes related to steatosis, fibrosis and inflammation. Conclusions: We provide a novel murine model for early-stage fatty liver disease suitable for drug testing and investigation of pathophysiology.
Collapse
|
84
|
Wang Q, Gu T, Ma L, Bu S, Zhou W, Mao G, Wang LL, Guo Y, Lai D. Efficient iron utilization compensates for loss of extracellular matrix of ovarian cancer spheroids. Free Radic Biol Med 2021; 164:369-380. [PMID: 33450374 DOI: 10.1016/j.freeradbiomed.2021.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 12/08/2020] [Accepted: 01/01/2021] [Indexed: 01/10/2023]
Abstract
Metastasis is the major cause of death in women with advanced ovarian cancer. Epithelial ovarian cancer cells can dissociate directly from extracellular matrix (ECM) and form spheroids to spread through the peritoneal cavity. Loss of ECM hinders the survival of ECM-detached epithelial cells. It is still largely unknown how ovarian cancer spheroids maintain their viability after loss of ECM. We find that spheroids derived either from ovarian cancer ascites or cell lines are iron-replete. In accordance with iron-replete condition, proteins involved in iron uptake, transport and storage including divalent metal ion transporter 1 (DMT1), transferrin receptor 1 (TFR1), ferritin, poly(rC)-binding proteins 1 and 2 (PCBP1 and 2) and nuclear factor E2-related factor 2 (NRF2) all increase in ovarian cancer spheroids. Genes linking iron homeostasis and lipid metabolism including stearoyl coenzyme A desaturase 1 (SCD1) are up-regulated in ovarian cancer spheroids. The product of SCD1 oleic acid can restore the viability of ovarian cancer spheroids inhibited by deprivation of iron. Extracellular signal-regulated kinase (ERK) activation contributes to autophagy activation in ovarian cancer spheroids. Impairment of autophagy by U0126 or Olaparib results in lysosomal iron accumulation and decrease of the cytosolic labile iron pool, leading to reduction of SCD1, lipid level and cell viability. Combination of U0126 and Olaparib has synergistic cytotoxicity toward ovarian cancer spheroids. Our findings reveal that ovarian cancer spheroids develop efficient iron utilization system to survive. Targeting iron utilization in ovarian cancer spheroids may have the potential to become new treatment strategies for ovarian cancer metastasis.
Collapse
Affiliation(s)
- Qian Wang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, PR China; Shanghai Municipal Key Clinical Specialty, Shanghai, 200030, PR China.
| | - Tingting Gu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Li Ma
- Zhongshan Hospital, Fudan University, Shanghai, 200030, PR China
| | - Shixia Bu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Wenjing Zhou
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Guoping Mao
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Lu-Lu Wang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Ying Guo
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Dongmei Lai
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, 200030, PR China; Shanghai Municipal Key Clinical Specialty, Shanghai, 200030, PR China.
| |
Collapse
|
85
|
Mechanisms of resistance to chemotherapy in non-small cell lung cancer. Arch Pharm Res 2021; 44:146-164. [PMID: 33608812 DOI: 10.1007/s12272-021-01312-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/16/2021] [Indexed: 02/07/2023]
Abstract
Non-small cell lung cancer (NSCLC), which represents 80-85% of lung cancer cases, is one of the leading causes of human death worldwide. The majority of patients undergo an intensive and invasive treatment regimen, which may include radiotherapy, chemotherapy, targeted therapy, immunotherapy, or a combination of these, depending on disease stage and performance status. Despite advances in therapeutic regimens, the 5-year survival of NSCLC is approximately 20-30%, largely due to diagnosis at advanced stages. Conventional chemotherapy is still the standard treatment option for patients with NSCLC, especially those with advanced disease. However, the emergence of resistance to chemotherapeutic agents (chemoresistance) poses a significant obstacle to the management of patients with NSCLC. Therefore, to develop efficacious chemotherapeutic approaches for NSCLC, it is necessary to understand the mechanisms underlying chemoresistance. Several mechanisms are known to mediate chemoresistance. These include altered cellular targets for chemotherapy, decreased cellular drug concentrations, blockade of chemotherapy-induced cell cycle arrest and apoptosis, acquisition of epithelial-mesenchymal transition and cancer stem cell-like phenotypes, deregulated expression of microRNAs, epigenetic modulation, and the interaction with tumor microenvironments. In this review, we summarize the mechanisms underlying chemoresistance and tumor recurrence in NSCLC and discuss potential strategies to avoid or overcome chemoresistance.
Collapse
|
86
|
Boldarine VT, Joyce E, Pedroso AP, Telles MM, Oyama LM, Bueno AA, Ribeiro EB. Oestrogen replacement fails to fully revert ovariectomy-induced changes in adipose tissue monoglycerides, diglycerides and cholesteryl esters of rats fed a lard-enriched diet. Sci Rep 2021; 11:3841. [PMID: 33589704 PMCID: PMC7884784 DOI: 10.1038/s41598-021-82837-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 01/22/2021] [Indexed: 01/09/2023] Open
Abstract
Menopause may be accompanied by abdominal obesity and inflammation, conditions accentuated by high-fat intake, especially of saturated fat (SFA)-rich diets. We investigated the consequences of high-SFA intake on the fatty acid (FA) profile of monoglycerides, diglycerides and cholesteryl esters from retroperitoneal white adipose tissue (RET) of rats with ovariectomy-induced menopause, and the effect of oestradiol replacement. Wistar rats were either ovariectomized (Ovx) or sham operated (Sham) and fed either standard chow (C) or lard-enriched diet (L) for 12 weeks. Half of the Ovx rats received 17β-oestradiol replacement (Ovx + E2). Body weight and food intake were measured weekly. RET neutral lipids were chromatographically separated and FAs analysed by gas chromatography. Ovariectomy alone increased body weight, feed efficiency, RET mass, leptin and insulin levels, leptin/adiponectin ratio, HOMA-IR and HOMA-β indexes. OvxC + E2 showed attenuation in nearly all blood markers. HOMA-β index was restored in OvxL + E2. OvxC showed significantly disturbed SFA and polyunsaturated FA (PUFA) profile in RET cholesteryl esters (CE). OvxC also showed increased monounsaturated FA (MUFA) in the monoglyceride diglyceride (Mono-Di) fraction. Similar changes were not observed in OvxL, although increased SFA and decreased PUFA was observed in Mono-Di. Overall, HRT was only partially able to revert changes induced by ovariectomy. There appears to be increased mobilization of essential FA in Ovx via CE, which is a dynamic lipid species. The same results were not found in Mono-Di, which are more inert. HRT may be helpful to preserve FA profile in visceral fat, but possibly not wholly sufficient in reverting the metabolic effects induced by menopause.
Collapse
Affiliation(s)
- Valter Tadeu Boldarine
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil.
| | - Ellen Joyce
- Department of Biological Sciences, College of Health, Life and Environmental Sciences, University of Worcester, Worcester, UK
| | - Amanda Paula Pedroso
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| | - Mônica Marques Telles
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| | - Lila Missae Oyama
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| | - Allain Amador Bueno
- Department of Biological Sciences, College of Health, Life and Environmental Sciences, University of Worcester, Worcester, UK
| | - Eliane Beraldi Ribeiro
- Departamento de Fisiologia, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Botucatu 862, 2º andar, Vila Clementino, São Paulo, SP, 04023-062, Brasil
| |
Collapse
|
87
|
Cariello M, Piccinin E, Moschetta A. Transcriptional Regulation of Metabolic Pathways via Lipid-Sensing Nuclear Receptors PPARs, FXR, and LXR in NASH. Cell Mol Gastroenterol Hepatol 2021; 11:1519-1539. [PMID: 33545430 PMCID: PMC8042405 DOI: 10.1016/j.jcmgh.2021.01.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease comprises a wide spectrum of liver injuries from simple steatosis to steatohepatitis and cirrhosis. Nonalcoholic steatohepatitis (NASH) is defined when liver steatosis is associated with inflammation, hepatocyte damage, and fibrosis. A genetic predisposition and environmental insults (ie, dietary habits, obesity) are putatively responsible for NASH progression. Here, we present the impact of the lipid-sensing nuclear receptors in the pathogenesis and treatment of NASH. In detail, we discuss the pros and cons of the putative transcriptional action of the fatty acid sensors (peroxisome proliferator-activated receptors), the bile acid sensor (farnesoid X receptor), and the oxysterol sensor (liver X receptors) in the pathogenesis and bona fide treatment of NASH.
Collapse
Affiliation(s)
- Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy
| | - Elena Piccinin
- Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro," Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro," Bari, Italy; National Institute for Biostructures and Biosystems (INBB), Rome, Italy; Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) Istituto Tumori Giovanni Paolo II, Bari, Italy.
| |
Collapse
|
88
|
Oatman N, Dasgupta N, Arora P, Choi K, Gawali MV, Gupta N, Parameswaran S, Salomone J, Reisz JA, Lawler S, Furnari F, Brennan C, Wu J, Sallans L, Gudelsky G, Desai P, Gebelein B, Weirauch MT, D'Alessandro A, Komurov K, Dasgupta B. Mechanisms of stearoyl CoA desaturase inhibitor sensitivity and acquired resistance in cancer. SCIENCE ADVANCES 2021; 7:eabd7459. [PMID: 33568479 PMCID: PMC7875532 DOI: 10.1126/sciadv.abd7459] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/22/2020] [Indexed: 05/22/2023]
Abstract
The lipogenic enzyme stearoyl CoA desaturase (SCD) plays a key role in tumor lipid metabolism and membrane architecture. SCD is often up-regulated and a therapeutic target in cancer. Here, we report the unexpected finding that median expression of SCD is low in glioblastoma relative to normal brain due to hypermethylation and unintentional monoallelic co-deletion with phosphatase and tensin homolog (PTEN) in a subset of patients. Cell lines from this subset expressed undetectable SCD, yet retained residual SCD enzymatic activity. Unexpectedly, these lines evolved to survive independent of SCD through unknown mechanisms. Cell lines that escaped such genetic and epigenetic alterations expressed higher levels of SCD and were highly dependent on SCD for survival. Last, we identify that SCD-dependent lines acquire resistance through a previously unknown FBJ murine osteosarcoma viral oncogene homolog B (FOSB)-mediated mechanism. Accordingly, FOSB inhibition blunted acquired resistance and extended survival of tumor-bearing mice treated with SCD inhibitor.
Collapse
Affiliation(s)
- Nicole Oatman
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nupur Dasgupta
- Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Priyanka Arora
- College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Kwangmin Choi
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Mruniya V Gawali
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Nishtha Gupta
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sreeja Parameswaran
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Joseph Salomone
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Julie A Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sean Lawler
- Neurosurgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Frank Furnari
- Ludwig Institute of Cancer Research, University of California, San Diego, CA, USA
| | | | - Jianqiang Wu
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Larry Sallans
- Department of Chemistry, University of Cincinnati, Cincinnati, OH, USA
| | - Gary Gudelsky
- College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Pankaj Desai
- College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| | - Brian Gebelein
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
- Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kakajan Komurov
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Biplab Dasgupta
- Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
- University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
89
|
Aramchol downregulates stearoyl CoA-desaturase 1 in hepatic stellate cells to attenuate cellular fibrogenesis. JHEP Rep 2021; 3:100237. [PMID: 34151243 PMCID: PMC8189934 DOI: 10.1016/j.jhepr.2021.100237] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 01/13/2021] [Accepted: 01/17/2021] [Indexed: 02/07/2023] Open
Abstract
Background & Aims Aramchol is a fatty acid-bile acid conjugate that reduces liver fat content and is being evaluated in a phase III clinical trial for non-alcoholic steatohepatitis (NASH). Aramchol attenuates NASH in mouse models and decreases steatosis by downregulating the fatty acid synthetic enzyme stearoyl CoA desaturase 1 (SCD1) in hepatocytes. Although hepatic stellate cells (HSCs) also store lipids as retinyl esters, the impact of Aramchol in this cell type is unknown. Methods We investigated the effects of Aramchol on a human HSC line (LX-2), primary human HSCs (phHSCs), and primary human hepatocytes (phHeps). Results In LX-2 and phHSCs, 10 μM Aramchol significantly reduced SCD1 mRNA while inducing PPARG (PPARγ) mRNA, with parallel changes in the 2 proteins; ACTA2, COL1A1, β-PDGFR (bPDGFR) mRNAs were also significantly reduced in LX-2. Secretion of collagen 1 (Col1α1) was inhibited by 10 μM Aramchol. SCD1 knockdown in LX-2 cells phenocopied the effect of Aramchol by reducing fibrogenesis, and addition of Aramchol to these cells did not rescue fibrogenic gene expression. Conversely, in LX-2 overexpressing SCD1, Aramchol no longer suppressed fibrogenic gene expression. The drug also induced genes in LX-2 that promote cholesterol efflux and inhibited ACAT2, which catalyses cholesterol synthesis. In phHeps, Aramchol also reduced SCD1 and increased PPARG mRNA expression. Conclusions Aramchol downregulates SCD1 and elevates PPARG in HSCs, reducing COL1A1 and ACTA2 mRNAs and COL1A1 secretion. These data suggest a direct inhibitory effect of Aramchol in HSCs through SCD1 inhibition, as part of a broader impact on both fibrogenic genes as well as mediators of cholesterol homeostasis. These findings illustrate novel mechanisms of Aramchol activity, including potential antifibrotic activity in patients with NASH and fibrosis. Lay summary In this study, we have explored the potential activity of Aramchol, a drug currently in clinical trials for fatty liver disease, in blocking fibrosis, or scarring, by hepatic stellate cells, the principal collagen-producing (i.e. fibrogenic) cell type in liver injury. In both isolated human hepatic stellate cells and in a human hepatic stellate cell line, the drug suppresses the key fat-producing enzyme, stearoyl CoA desaturase 1 (SCD1), which leads to reduced expression of genes and proteins associated with hepatic fibrosis, while inducing the protective gene, PPARγ. The drug loses activity when SCD1 is already reduced by gene knockdown, reinforcing the idea that inhibition of SCD1 is a main mode of activity for Aramchol. These findings strengthen the rationale for testing Aramchol in patients with NASH. The antifibrotic activity of Aramchol was assessed in human hepatic stellate cells (HSCs). Aramchol reduces fibrogenic gene expression by inhibiting SCD1 and inducing PPARγ. Aramchol inhibits pathways that increase HSC cholesterol content. The antifibrotic activity of Aramchol reinforces its potential efficacy in human NASH.
Collapse
Key Words
- ABCA1, ATP-binding cassette transporter 1
- EMT, epithelial-mesenchymal-transition
- Fatty liver disease
- Fibrosis
- GSEA, gene set enrichment analysis
- GSH, glutathione
- GSSG, glutathione disulfide
- HRP, horse radish peroxidase
- HSC, hepatic stellate cell
- Hepatic fibrosis
- Hh, Hedgehog
- MCD, methionine-choline depleted diet
- MMP-2, matrix metalloproteinase 2
- MUFAs, monounsaturated fatty acids
- NAFLD, non-alcoholic fatty liver disease
- NASH, non-alcoholic steatohepatitis
- Non-alcoholic steatohepatitis
- PDMS, polydimethylsiloxane
- PPAR, peroxisome proliferator-activated receptor
- SCD1, stearoyl CoA-desaturase 1
- SMA, smooth muscle actin
- TAA, thioacetamide
- phHSCs, primary human hepatic stellate cells
- phHeps, primary human hepatocytes
- siRNA, small inhibitory RNA
Collapse
|
90
|
Romano A, Friuli M, Del Coco L, Longo S, Vergara D, Del Boccio P, Valentinuzzi S, Cicalini I, Fanizzi FP, Gaetani S, Giudetti AM. Chronic Oleoylethanolamide Treatment Decreases Hepatic Triacylglycerol Level in Rat Liver by a PPARγ/SREBP-Mediated Suppression of Fatty Acid and Triacylglycerol Synthesis. Nutrients 2021; 13:394. [PMID: 33513874 PMCID: PMC7910994 DOI: 10.3390/nu13020394] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/18/2021] [Accepted: 01/23/2021] [Indexed: 12/14/2022] Open
Abstract
Oleoylethanolamide (OEA) is a naturally occurring bioactive lipid belonging to the family of N-acylethanolamides. A variety of beneficial effects have been attributed to OEA, although the greater interest is due to its potential role in the treatment of obesity, fatty liver, and eating-related disorders. To better clarify the mechanism of the antiadipogenic effect of OEA in the liver, using a lipidomic study performed by 1H-NMR, LC-MS/MS and thin-layer chromatography analyses we evaluated the whole lipid composition of rat liver, following a two-week daily treatment of OEA (10 mg kg-1 i.p.). We found that OEA induced a significant reduction in hepatic triacylglycerol (TAG) content and significant changes in sphingolipid composition and ceramidase activity. We associated the antiadipogenic effect of OEA to decreased activity and expression of key enzymes involved in fatty acid and TAG syntheses, such as acetyl-CoA carboxylase, fatty acid synthase, diacylglycerol acyltransferase, and stearoyl-CoA desaturase 1. Moreover, we found that both SREBP-1 and PPARγ protein expression were significantly reduced in the liver of OEA-treated rats. Our findings add significant and important insights into the molecular mechanism of OEA on hepatic adipogenesis, and suggest a possible link between the OEA-induced changes in sphingolipid metabolism and suppression of hepatic TAG level.
Collapse
Affiliation(s)
- Adele Romano
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (A.R.); (M.F.); (S.G.)
| | - Marzia Friuli
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (A.R.); (M.F.); (S.G.)
| | - Laura Del Coco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Prov.le Lecce-Monteroni, 73100 Lecce, Italy; (L.D.C.); (S.L.); (D.V.)
| | - Serena Longo
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Prov.le Lecce-Monteroni, 73100 Lecce, Italy; (L.D.C.); (S.L.); (D.V.)
| | - Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Prov.le Lecce-Monteroni, 73100 Lecce, Italy; (L.D.C.); (S.L.); (D.V.)
| | - Piero Del Boccio
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (P.D.B.); (S.V.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy;
| | - Silvia Valentinuzzi
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy; (P.D.B.); (S.V.)
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy;
| | - Ilaria Cicalini
- Center for Advanced Studies and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy;
- Department of Medicine and Aging Science, University “G. d’Annunzio” of Chieti-Pescara, 66100 Chieti, Italy
| | - Francesco P. Fanizzi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Prov.le Lecce-Monteroni, 73100 Lecce, Italy; (L.D.C.); (S.L.); (D.V.)
| | - Silvana Gaetani
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (A.R.); (M.F.); (S.G.)
| | - Anna M. Giudetti
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Prov.le Lecce-Monteroni, 73100 Lecce, Italy; (L.D.C.); (S.L.); (D.V.)
| |
Collapse
|
91
|
Ye S, Matthan NR, Lamon-Fava S, Aguilar GS, Turner JR, Walker ME, Chai Z, Lakshman S, Urban JF, Lichtenstein AH. Western and heart healthy dietary patterns differentially affect the expression of genes associated with lipid metabolism, interferon signaling and inflammation in the jejunum of Ossabaw pigs. J Nutr Biochem 2020; 90:108577. [PMID: 33388349 PMCID: PMC8982565 DOI: 10.1016/j.jnutbio.2020.108577] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022]
Abstract
Diet quality and statin therapy are established modulators of coronary artery disease (CAD) progression, but their effect on the gastrointestinal tract and subsequent sequelae that could affect CAD progression are relatively unexplored. To address this gap, Ossabaw pigs (N = 32) were randomly assigned to receive isocaloric amounts of a Western-type diet (WD; high in saturated fat, refined carbohydrate, and cholesterol, and low in fiber) or a heart healthy-type diet (HHD; high in unsaturated fat, whole grains, fruits and vegetables, supplemented with fish oil, and low in cholesterol), with or without atorvastatin, for 6 months. At the end of the study, RNA sequencing with 100 base pair single end reads on NextSeq 500 platform was conducted in isolated pig jejunal mucosa. A two-factor edgeR analysis revealed that the dietary patterns resulted in three differentially expressed genes related to lipid metabolism (SCD, FADS1, and SQLE). The expression of these genes was associated with cardiometabolic risk factors and atherosclerotic lesion severity. Subsequent gene enrichment analysis indicated the WD, compared to the HHD, resulted in higher interferon signaling and inflammation, with some of these genes being significantly associated with serum TNF-α and/or hsCRP concentrations, but not atherosclerotic lesion severity. No significant effect of atorvastatin therapy on gene expression, nor its interaction with dietary patterns, was identified. In conclusion, Western and heart healthy-type dietary patterns differentially affect the expression of genes associated with lipid metabolism, interferon signaling, and inflammation in the jejunum of Ossabaw pigs.
Collapse
Affiliation(s)
- Shumao Ye
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Nirupa R Matthan
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Stefania Lamon-Fava
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA
| | - Gloria Solano Aguilar
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD, USA
| | - Jerrold R Turner
- Laboratory of Mucosal Barrier Pathobiology, Department of Pathology, Brigham and Woman's Hospital and Harvard Medical School, Boston, MA, USA
| | - Maura E Walker
- Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA, USA
| | - Zhi Chai
- Intercollege Graduate Degree Program in Physiology, Department of Nutritional Science, Pennsylvania State University, University Park, PA, USA
| | - Sukla Lakshman
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD, USA
| | - Joseph F Urban
- USDA, ARS, Beltsville Human Nutrition Research Center, Diet Genomics and Immunology Laboratory, Beltsville, MD, USA
| | - Alice H Lichtenstein
- Cardiovascular Nutrition Laboratory, Jean Mayer USDA Human Nutrition Research Center on Aging; Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, USA.
| |
Collapse
|
92
|
Bogie JFJ, Grajchen E, Wouters E, Corrales AG, Dierckx T, Vanherle S, Mailleux J, Gervois P, Wolfs E, Dehairs J, Van Broeckhoven J, Bowman AP, Lambrichts I, Gustafsson JÅ, Remaley AT, Mulder M, Swinnen JV, Haidar M, Ellis SR, Ntambi JM, Zelcer N, Hendriks JJA. Stearoyl-CoA desaturase-1 impairs the reparative properties of macrophages and microglia in the brain. J Exp Med 2020; 217:133840. [PMID: 32097464 PMCID: PMC7201924 DOI: 10.1084/jem.20191660] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/12/2019] [Accepted: 01/24/2020] [Indexed: 12/15/2022] Open
Abstract
Failure of remyelination underlies the progressive nature of demyelinating diseases such as multiple sclerosis. Macrophages and microglia are crucially involved in the formation and repair of demyelinated lesions. Here we show that myelin uptake temporarily skewed these phagocytes toward a disease-resolving phenotype, while sustained intracellular accumulation of myelin induced a lesion-promoting phenotype. This phenotypic shift was controlled by stearoyl-CoA desaturase-1 (SCD1), an enzyme responsible for the desaturation of saturated fatty acids. Monounsaturated fatty acids generated by SCD1 reduced the surface abundance of the cholesterol efflux transporter ABCA1, which in turn promoted lipid accumulation and induced an inflammatory phagocyte phenotype. Pharmacological inhibition or phagocyte-specific deficiency of Scd1 accelerated remyelination ex vivo and in vivo. These findings identify SCD1 as a novel therapeutic target to promote remyelination.
Collapse
Affiliation(s)
- Jeroen F J Bogie
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Elien Grajchen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Elien Wouters
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Aida Garcia Corrales
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Tess Dierckx
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jo Mailleux
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Pascal Gervois
- Department of Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Esther Wolfs
- Department of Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jonas Dehairs
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, University of Leuven, Leuven, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Andrew P Bowman
- The Maastricht MultiModal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - Ivo Lambrichts
- Department of Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Alan T Remaley
- Lipoprotein Metabolism Laboratory, Translational Vascular Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Monique Mulder
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Johannes V Swinnen
- Department of Oncology, Laboratory of Lipid Metabolism and Cancer, Leuven Cancer Institute, University of Leuven, Leuven, Belgium
| | - Mansour Haidar
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Shane R Ellis
- The Maastricht MultiModal Molecular Imaging Institute, Division of Imaging Mass Spectrometry, Maastricht University, Maastricht, Netherlands
| | - James M Ntambi
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI.,Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI
| | - Noam Zelcer
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jerome J A Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
93
|
Yegorova S, Yegorov O, Ferreira LF. RNA-sequencing reveals transcriptional signature of pathological remodeling in the diaphragm of rats after myocardial infarction. Gene 2020; 770:145356. [PMID: 33333219 DOI: 10.1016/j.gene.2020.145356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/11/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
The diaphragm is the main inspiratory muscle, and the chronic phase post-myocardial infarction (MI) is characterized by diaphragm morphological, contractile, and metabolic abnormalities. However, the mechanisms of diaphragm weakness are not fully understood. In the current study, we aimed to identify the transcriptome changes associated with diaphragm abnormalities in the chronic stage MI. We ligated the left coronary artery to cause MI in rats and performed RNA-sequencing (RNA-Seq) in diaphragm samples 16 weeks post-surgery. The sham group underwent thoracotomy and pericardiotomy but no artery ligation. We identified 112 differentially expressed genes (DEGs) out of a total of 9664 genes. Myocardial infarction upregulated and downregulated 42 and 70 genes, respectively. Analysis of DEGs in the framework of skeletal muscle-specific biological networks suggest remodeling in the neuromuscular junction, extracellular matrix, sarcomere, cytoskeleton, and changes in metabolism and iron homeostasis. Overall, the data are consistent with pathological remodeling of the diaphragm and reveal potential biological targets to prevent diaphragm weakness in the chronic stage MI.
Collapse
Affiliation(s)
- Svetlana Yegorova
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA.
| | - Oleg Yegorov
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA.
| | - Leonardo F Ferreira
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
94
|
Yu G, Yang Z, Peng T, Lv Y. Circular RNAs: Rising stars in lipid metabolism and lipid disorders. J Cell Physiol 2020; 236:4797-4806. [PMID: 33275299 DOI: 10.1002/jcp.30200] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/03/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023]
Abstract
The underlying mechanisms of circular RNAs (circRNAs) in lipid metabolism regulation and the pathogenesis of lipid disorder diseases are clarified in this review. circRNAs are produced from host genes by back splicing and are mainly degraded by RNase L. circRNAs act as molecular sponges or scaffolds that bind with microRNAs or proteins and thus affect the intracorporeal processes of lipid metabolism. CircRNA_11897 and circSAMD4A facilitated adipogenesis while circH19 and circRNA_26852 accelerated adipolysis in adipose tissue. CircSAMD4A promoted the differentiation of preadipocytes, but circH19 and circFUT10 inhibited this differentiation. CircFUT10 also promoted the proliferation of preadipocytes. CiRS-133 fostered the browning of white adipose tissue. CircACC1, circRNA_021412, circRNA_0046366, and circRNA_0046367 promoted the mitochondrial β-oxidation of fatty acids in hepatocytes. CircRNA_021412 suppressed the synthesis of triglycerides in hepatocytes. CircScd1 inhibited hepatic lipid droplet formation. circ_0092317, circ_0003546, circ_0028198, circ_0092317, and circACC1 probably reduced cholesterol efflux from macrophages. circ_0037251 likely promoted lipid accumulation and inhibited lipophagy in macrophages. circRNAs participate in lipid metabolism regulation and affect the development of lipid disorder diseases.
Collapse
Affiliation(s)
- Guangli Yu
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhou Yang
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Tianhong Peng
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Yuncheng Lv
- Clinical Anatomy & Reproductive Medicine Application Institute, Hengyang Medical College, University of South China, Hengyang, Hunan, China.,Institute of Basic Medical Sciences & Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin, Guangxi, China
| |
Collapse
|
95
|
Ganbold M, Ferdousi F, Arimura T, Tominaga K, Isoda H. New Amphiphilic Squalene Derivative Improves Metabolism of Adipocytes Differentiated From Diabetic Adipose-Derived Stem Cells and Prevents Excessive Lipogenesis. Front Cell Dev Biol 2020; 8:577259. [PMID: 33251210 PMCID: PMC7672044 DOI: 10.3389/fcell.2020.577259] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022] Open
Abstract
Squalene (Sq) is a natural compound, found in various plant oils, algae, and larger quantity in deep-sea shark liver. It is also known as an intermediate of cholesterol synthesis in plants and animals including humans. Although evidences demonstrated its antioxidant, anticancer, hypolipidemic, and hepatoprotective and cardioprotective effects, its biological effects in cellular function might have been underestimated because of the water-insoluble property. To overcome this hydrophobicity, we synthesized new amphiphilic Sq derivative (HH-Sq). On the other hand, adipose-derived stem cells (ASCs) are a valuable source in regenerative medicine for its ease of accessibility and multilineage differentiation potential. Nevertheless, impaired cellular functions of ASCs derived from diabetic donor have still been debated controversially. In this study, we explored the effect of the HH-Sq in comparison to Sq on the adipocyte differentiation of ASCs obtained from subjects with type 2 diabetes. Gene expression profile by microarray analysis at 14 days of adipogenic differentiation revealed that HH-Sq induced more genes involved in intracellular signaling processes, whereas Sq activated more transmembrane receptor pathway-related genes. In addition, more important number of down-regulated and up-regulated genes by Sq and HH-Sq were not overlapped, suggesting the compounds might not only have difference in their chemical property but also potentially exert different biological effects. Both Sq and HH-Sq improved metabolism of adipocytes by enhancing genes associated with energy homeostasis and insulin sensitivity, SIRT1, PRKAA2, and IRS1. Interestingly, Sq increased significantly early adipogenic markers and lipogenic gene expression such as PPARG, SREBF1, and CEBPA, but not HH-Sq. As a consequence, smaller and fewer lipid droplet formation was observed in HH-Sq-treated adipocytes. Based on our findings, we report that both Sq and HH-Sq improved adipocyte metabolism, but only HH-Sq prevented excessive lipogenesis without abrogating adipocyte differentiation. The beneficial effect of HH-Sq provides an importance of synthesized derivatives from a natural compound with therapeutic potentials in the application of cell therapies.
Collapse
Affiliation(s)
- Munkhzul Ganbold
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), Tsukuba, Ibaraki, Japan
| | - Farhana Ferdousi
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), Tsukuba, Ibaraki, Japan.,Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Takashi Arimura
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), Tsukuba, Ibaraki, Japan
| | - Kenichi Tominaga
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), Tsukuba, Ibaraki, Japan
| | - Hiroko Isoda
- National Institute of Advanced Industrial Science and Technology (AIST)-University of Tsukuba Open Innovation Laboratory for Food and Medicinal Resource Engineering (FoodMed-OIL), Tsukuba, Ibaraki, Japan.,Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, Tsukuba, Ibaraki, Japan.,Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
96
|
Liu X, Zhao K, Jing N, Zhao Y, Yang X. EGCG regulates fatty acid metabolism of high-fat diet-fed mice in association with enrichment of gut Akkermansia muciniphila. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104261] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
97
|
Zhao J, Lin X, Meng D, Zeng L, Zhuang R, Huang S, Lv W, Hu J. Nrf2 Mediates Metabolic Reprogramming in Non-Small Cell Lung Cancer. Front Oncol 2020; 10:578315. [PMID: 33324555 PMCID: PMC7726415 DOI: 10.3389/fonc.2020.578315] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/22/2020] [Indexed: 01/14/2023] Open
Abstract
Nuclear factor erythroid-2–related factor-2 (NFE2L2/Nrf2) is a transcription factor that regulates the expression of antioxidant genes. Both Kelch-like ECH-associated protein 1 (Keap1) mutations and Nrf2 mutations contribute to the activation of Nrf2 in non-small cell lung cancer (NSCLC). Nrf2 activity is associated with poor prognosis in NSCLC. Metabolic reprogramming represents a cancer hallmark. Increasing studies reveal that Nrf2 activation promotes metabolic reprogramming in cancer. In this review, we discuss the underlying mechanisms of Nrf2-mediated metabolic reprogramming and elucidate its role in NSCLC. Inhibition of Nrf2 can alter metabolic processes, thus suppress tumor growth, prevent metastasis, and increase sensitivity to chemotherapy in NSCLC. In conclusion, Nrf2 may serve as a therapeutic target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Jiangang Zhao
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xu Lin
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Di Meng
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liping Zeng
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Runzhou Zhuang
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Sha Huang
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wang Lv
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
98
|
Piccinin E, Cariello M, Moschetta A. Lipid metabolism in colon cancer: Role of Liver X Receptor (LXR) and Stearoyl-CoA Desaturase 1 (SCD1). Mol Aspects Med 2020; 78:100933. [PMID: 33218679 DOI: 10.1016/j.mam.2020.100933] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 11/01/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is one of the most commonly occurring cancers worldwide. Although several genetic alterations have been associated with CRC onset and progression, nowadays the reprogramming of cellular metabolism has been recognized as a fundamental step of the carcinogenic process. Intestinal tumor cells frequently display an aberrant activation of lipid metabolism. Indeed, to satisfy the growing needs of a continuous proliferation, cancer cells can either increase the uptake of exogenous lipids or upregulate the endogenous lipogenesis and cholesterol synthesis. Therefore, strategies aimed at limiting lipid accumulation are now under development in order to counteract malignancies. Two major players of lipids metabolism have been so far identified for their contribution to CRC development: the nuclear receptor Liver X Receptor (LXRs) and the enzyme Stearoyl-CoA Desaturase 1 (SCD1). Whereas LXR is mainly recognized for its role as a cholesterol sensor, finally promoting the loss of cellular cholesterol and whole-body homeostasis, SCD1 acts as the major regulator of new fatty acids, finely tuning the monounsaturated fatty acids (MUFA) to saturated fatty acids (SFA) ratio. Intriguingly, SCD1 is directly regulated by LXRs. Despite LXRs agonists have elicited great interest as a promising therapeutic target for cancer, LXR's ability to induce SCD1 and new fatty acids synthesis represent a major obstacle in the development of new effective treatments. Thus, further investigations are required to fully dissect the concomitant modulation of both players, to develop specific therapies aimed at blocking intestinal cancer cells proliferation, eventually counteracting CRC progression.
Collapse
Affiliation(s)
- Elena Piccinin
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Bari, Italy; Department of Basic Medical Sciences, Neurosciences and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | - Marica Cariello
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Bari, Italy; INBB, National Institute for Biostructures and Biosystems, Rome, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Bari, Italy; INBB, National Institute for Biostructures and Biosystems, Rome, Italy; National Cancer Center, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy.
| |
Collapse
|
99
|
Abstract
Increasing evidence suggests a significant role for microbiota dependent metabolites and co-metabolites, acting as aryl hydrocarbon receptor (AHR) ligands, to facilitate bidirectional communication between the host and the microbiota and thus modulate physiology. Such communication is particularly evident within the gastrointestinal tract. Through binding to or activating the AHR, these metabolites play fundamental roles in various physiological processes and likely contribute to the maintenance of intestinal homeostasis. In recent years, tryptophan metabolites were screened to identify physiologically relevant AHR ligands or activators. The discovery of specific microbiota-derived indole-based metabolites as AHR ligands may provide insight concerning how these metabolites affect interactions between gut microbiota and host intestinal homeostasis and how this relates to chronic GI disease and overall health. A greater understanding of the mechanisms that modulate the production of such metabolites and associated AHR activity may be utilized to effectively treat inflammatory diseases and promote human health. Here, we review microbiota-derived AHR ligands generated from tryptophan that modulate host-gut microbiota interactions and discuss possible intervention strategies for potential therapies in the future.
Collapse
Affiliation(s)
- Fangcong Dong
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | - Gary H. Perdew
- Department of Veterinary and Biomedical Sciences and the Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA,CONTACT Gary H. Perdew The Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA16802, USA
| |
Collapse
|
100
|
Zhang Y, Zheng Y, Wang X, Qiu J, Liang C, Cheng G, Wang H, Zhao C, Yang W, Zan L, Li A. Bovine Stearoyl-CoA Desaturase 1 Promotes Adipogenesis by Activating the PPARγ Receptor. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:12058-12066. [PMID: 33052678 DOI: 10.1021/acs.jafc.0c05147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stearoyl-CoA desaturase 1 (SCD1) is a rate-limiting enzyme that mainly catalyzes the saturated fatty acids (SFAs) into the monounsaturated fatty acids (MUFAs). The expression level of SCD1 is positively correlated with the marbling score. However, the functional mechanism of SCD1 in adipogenesis is still unclear. In this study, we identified SCD1 as highly expressed in subcutaneous and visceral fat, peaking at 2 days after differentiation in bovine stromal vascular fraction (SVF) cells. When the SCD1 was overexpressed in bovine SVF cells, lipid droplets accumulation was increased from 142.46 ± 21.77 to 254.89 ± 11.75 μg/mg (P < 0.01). Further, the expression levels of FABP4, FASN, and ACCα were increased (P < 0.01), while the expression of PPARγ or C/EBPα was not changed at mRNA or protein level (P > 0.05). Dual-luciferase reporter assay showed that the activity of the PPARγ receptor was enhanced by 3.69 times (P < 0.01). Moreover, the contents of palmitoleate (C16:1) and oleate (C18:1) were significantly increased (P < 0.05). Furthermore, 100 μM exogenous oleate increased the lipid accumulation by 22.28 times (P < 0.01). These results suggest that oleate is probably a strong ligand of the PPARγ receptor to enhance adipogenesis.
Collapse
Affiliation(s)
- Yu Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Yan Zheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Xiaoyu Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Ju Qiu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Chengcheng Liang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Gong Cheng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Hongbao Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Chunping Zhao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Wucai Yang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Linsen Zan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| | - Anning Li
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
- National Beef Cattle Improvement Center, Northwest A&F University, Yangling, Shaanxi 712100, P. R. China
| |
Collapse
|