1
|
Sun F, Wang J, Yang Y, Dong QQ, Jia L, Hu W, Tao H, Lu C, Yang JJ. Epitranscriptomic regulation of lipid oxidation and liver fibrosis via ENPP1 mRNA m 6A modification. Cell Mol Life Sci 2024; 81:387. [PMID: 39249529 PMCID: PMC11383905 DOI: 10.1007/s00018-024-05420-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/15/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Dysregulated lipid oxidation occurs in several pathological processes characterized by cell proliferation and migration. Nonetheless, the molecular mechanism of lipid oxidation is not well appreciated in liver fibrosis, which is accompanied by enhanced fibroblast proliferation and migration. METHODS We investigated the causes and consequences of lipid oxidation in liver fibrosis using cultured cells, animal models, and clinical samples. RESULTS Increased ecto-nucleotide pyrophosphatase/phosphodiesterase (ENPP1) expression caused increased lipid oxidation, resulting in the proliferation and migration of hepatic stellate cells (HSCs) that lead to liver fibrosis, whereas fibroblast-specific ENPP1 knockout reversing these results. Elevated ENPP1 and N6-methyladenosine (m6A) levels were associated with high expression of Wilms tumor 1 associated protein (WTAP). Mechanistically, WTAP-mediated m6A methylation of the 3'UTR of ENPP1 mRNA and induces its translation dependent of YTH domain family proteins 1 (YTHDF1). Additionally, ENPP1 could interact with hypoxia inducible lipid droplet associated (HILPDA) directly; overexpression of ENPP1 further recruits HILPDA-mediated lipid oxidation, thereby promotes HSCs proliferation and migration, while inhibition of ENPP1 expression produced the opposite effect. Clinically, increased expression of WTAP, YTHDF1, ENPP1, and HILPDA, and increased m6A mRNA content, enhanced lipid oxidation, and increased collagen deposition in human liver fibrosis tissues. CONCLUSIONS We describe a novel mechanism in which WTAP catalyzes m6A methylation of ENPP1 in a YTHDF1-dependent manner to enhance lipid oxidation, promoting HSCs proliferation and migration and liver fibrosis.
Collapse
Affiliation(s)
- Feng Sun
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Juan Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Yang Yang
- Department of General Surgery, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215153, China
| | - Qi-Qi Dong
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Lin Jia
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hui Tao
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, People's Republic of China.
| | - Chao Lu
- First Affiliated Hospital, Anhui University of Science and Technology, Huainan, 232001, China.
| | - Jing-Jing Yang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
| |
Collapse
|
2
|
Khazaei-Koohpar H, Gholizadeh M, Hafezian SH, Esmaeili-Fard SM. Weighted single-step genome-wide association study for direct and maternal genetic effects associated with birth and weaning weights in sheep. Sci Rep 2024; 14:13120. [PMID: 38849438 PMCID: PMC11161479 DOI: 10.1038/s41598-024-63974-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 06/04/2024] [Indexed: 06/09/2024] Open
Abstract
Body weight is an important economic trait for sheep meat production, and its genetic improvement is considered one of the main goals in the sheep breeding program. Identifying genomic regions that are associated with growth-related traits accelerates the process of animal breeding through marker-assisted selection, which leads to increased response to selection. In this study, we conducted a weighted single-step genome-wide association study (WssGWAS) to identify potential candidate genes for direct and maternal genetic effects associated with birth weight (BW) and weaning weight (WW) in Baluchi sheep. The data used in this research included 13,408 birth and 13,170 weaning records collected at Abbas-Abad Baluchi Sheep Breeding Station, Mashhad-Iran. Genotypic data of 94 lambs genotyped by Illumina 50K SNP BeadChip for 54,241 markers were used. The proportion of variance explained by genomic windows was calculated by summing the variance of SNPs within 1 megabase (Mb). The top 10 window genomic regions explaining the highest percentages of additive and maternal genetic variances were selected as candidate window genomic regions associated with body weights. Our findings showed that for BW, the top-ranked genomic regions (1 Mb windows) explained 4.30 and 4.92% of the direct additive and maternal genetic variances, respectively. The direct additive genetic variance explained by the genomic window regions varied from 0.31 on chromosome 1 to 0.59 on chromosome 8. The highest (0.84%) and lowest (0.32%) maternal genetic variances were explained by genomic windows on chromosome 10 and 17, respectively. For WW, the top 10 genomic regions explained 6.38 and 5.76% of the direct additive and maternal genetic variances, respectively. The highest and lowest contribution of direct additive genetic variances were 1.37% and 0.42%, respectively, both explained by genomic regions on chromosome 2. For maternal effects on WW, the highest (1.38%) and lowest (0.41%) genetic variances were explained by genomic windows on chromosome 2. Further investigation of these regions identified several possible candidate genes associated with body weight. Gene ontology analysis using the DAVID database identified several functional terms, such as translation repressor activity, nucleic acid binding, dehydroascorbic acid transporter activity, growth factor activity and SH2 domain binding.
Collapse
Affiliation(s)
- Hava Khazaei-Koohpar
- Department of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University (SANRU), Sari, Iran
| | - Mohsen Gholizadeh
- Department of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University (SANRU), Sari, Iran.
| | - Seyed Hasan Hafezian
- Department of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University (SANRU), Sari, Iran
| | | |
Collapse
|
3
|
Smith DM, Liu BY, Wolfgang MJ. Rab30 facilitates lipid homeostasis during fasting. Nat Commun 2024; 15:4469. [PMID: 38796472 PMCID: PMC11127972 DOI: 10.1038/s41467-024-48959-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
To facilitate inter-tissue communication and the exchange of proteins, lipoproteins, and metabolites with the circulation, hepatocytes have an intricate and efficient intracellular trafficking system regulated by small Rab GTPases. Here, we show that Rab30 is induced in the mouse liver by fasting, which is amplified in liver-specific carnitine palmitoyltransferase 2 knockout mice (Cpt2L-/-) lacking the ability to oxidize fatty acids, in a Pparα-dependent manner. Live-cell super-resolution imaging and in vivo proximity labeling demonstrates that Rab30-marked vesicles are highly dynamic and interact with proteins throughout the secretory pathway. Rab30 whole-body, liver-specific, and Rab30; Cpt2 liver-specific double knockout (DKO) mice are viable with intact Golgi ultrastructure, although Rab30 deficiency in DKO mice suppresses the serum dyslipidemia observed in Cpt2L-/- mice. Corresponding with decreased serum triglyceride and cholesterol levels, DKO mice exhibit decreased circulating but not hepatic ApoA4 protein, indicative of a trafficking defect. Together, these data suggest a role for Rab30 in the selective sorting of lipoproteins to influence hepatocyte and circulating triglyceride levels, particularly during times of excessive lipid burden.
Collapse
Affiliation(s)
- Danielle M Smith
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Brian Y Liu
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Michael J Wolfgang
- Department of Physiology, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biological Chemistry, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
4
|
Li H, Seessle J, Staffer S, Tuma-Kellner S, Poschet G, Herrmann T, Chamulitrat W. FATP4 deletion in liver cells induces elevation of extracellular lipids via metabolic channeling towards triglycerides and lipolysis. Biochem Biophys Res Commun 2023; 687:149161. [PMID: 37931418 DOI: 10.1016/j.bbrc.2023.149161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/13/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
Evidence from mice with global deletion of fatty-acid transport protein4 (FATP4) indicates its role on β-oxidation and triglycerides (TG) metabolism. We reported that plasma glycerol and free fatty acids (FA) were increased in liver-specific Fatp4 deficient (L-FATP4-/-) mice under dietary stress. We hypothesized that FATP4 may mediate hepatocellular TG lipolysis. Here, we demonstrated that L-FATP4-/- mice showed an increase in these blood lipids, liver TG, and subcutaneous fat weights. We therefore studied TG metabolism in response to oleate treatment in two experimental models using FATP4-knockout HepG2 (HepKO) cells and L-FATP4-/- hepatocytes. Both FATP4-deificient liver cells showed a significant decrease in β-oxidation products by ∼30-35% concomitant with marked upregulation of CD36, FATP2, and FATP5 as well as lipoprotein microsomal-triglyceride-transfer protein genes. By using 13C3D5-glycerol, HepKO cells displayed an increase in metabolically labelled TG species which were further increased with oleate treatment. This increase was concomitant with a step-wise elevation of TG in cells and supernatants as well as the secretion of cholesterol very low-density and high-density lipoproteins. Upon analyzing TG lipolytic enzymes, both mutant liver cells showed marked upregulated expression of hepatic lipase, while that of hormone-sensitive lipase and adipose-triglyceride lipase was downregulated. Lipolysis measured by extracellular glycerol and free FA was indeed increased in mutant cells, and this event was exacerbated by oleate treatment. Taken together, FATP4 deficiency in liver cells led to a metabolic shift from β-oxidation towards lipolysis-directed TG and lipoprotein secretion, which is in line with an association of FATP4 polymorphisms with blood lipids.
Collapse
Affiliation(s)
- Huili Li
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, Hubei, China
| | - Jessica Seessle
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Simone Staffer
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, University of Heidelberg, 69120, Heidelberg, Germany
| | - Thomas Herrmann
- Westkuesten Hospital, Esmarchstraße 50, 25746, Heide, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
5
|
Wang S, Lu Y, Chi T, Zhang Y, Zhao Y, Guo H, Feng L. Identification of ferroptosis-related genes in type 2 diabetes mellitus based on machine learning. Immun Inflamm Dis 2023; 11:e1036. [PMID: 37904700 PMCID: PMC10566453 DOI: 10.1002/iid3.1036] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 09/05/2023] [Accepted: 09/17/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM), which has a high incidence and several harmful consequences, poses a severe danger to human health. Research on the function of ferroptosis in T2DM is increasing. This study uses bioinformatics techniques identify new diagnostic T2DM biomarkers associated with ferroptosis. METHODS To identify ferroptosis-related genes (FRGs) that are differentially expressed between T2DM patients and healthy individuals, we first obtained T2DM sequencing data and FRGs from the Gene Expression Omnibus (GEO) database and FerrDb database. Then, drug-gene interaction networks and competitive endogenous RNA (ceRNA) networks linked to the marker genes were built after marker genes were filtered by two machine learning algorithms (LASSO and SVM-RFE algorithms). Finally, to confirm the expression of marker genes, the GSE76895 dataset was utilized. The protein and RNA expression of some marker genes in T2DM and nondiabetic tissues was also examined by Western blotting, immunohistochemistry (IHC), immunofluorescence (IF) and quantitative real-time PCR (qRT-PCR). RESULTS We obtained 58 differentially expressed genes (DEGs) associated with ferroptosis. GO and KEGG enrichment analyses showed that these DEGs were significantly enriched in hypoxia and ferroptosis. Subsequently, eight marker genes (SCD, CD44, HIF1A, BCAT2, MTF1, HILPDA, NR1D2, and MYCN) were screened by LASSO and SVM-RFE machine learning algorithms, and a model was constructed based on these eight genes. This model also has high diagnostic power. In addition, based on these eight genes, we obtained 48 drugs and constructed a complex ceRNA network map. Finally, Western blotting, IHC, IF, and qRT-PCR results of clinical samples further confirmed the results of public databases. CONCLUSIONS The diagnosis and aetiology of T2DM can be greatly aided by eight FRGs, providing novel therapeutic avenues.
Collapse
Affiliation(s)
- Sen Wang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Yongpan Lu
- Department of Plastic Surgery, The First Clinical Medical College, Shandong University of Traditional Chinese MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalJinanShandongChina
| | - Tingting Chi
- Department of Acupuncture and RehabilitationThe Affiliated Qingdao Hai Ci Hospital of Qingdao University (West Hospital Area)QingdaoShandongChina
| | - Yixin Zhang
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Yuli Zhao
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Huimin Guo
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| | - Li Feng
- Department of Medical Ultrasound, Shandong Medicine and Health Key Laboratory of Abdominal Medical Imaging, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qian Foshan HospitalShandong First Medical UniversityJinanShandongChina
| |
Collapse
|
6
|
Deng L, Wu SA, Qi L, Kersten S. HILPDA is a lipotoxic marker in adipocytes that mediates the autocrine negative feedback regulation of triglyceride hydrolysis by fatty acids and alleviates cellular lipotoxic stress. Mol Metab 2023; 75:101773. [PMID: 37422000 PMCID: PMC10391665 DOI: 10.1016/j.molmet.2023.101773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/03/2023] [Accepted: 07/04/2023] [Indexed: 07/10/2023] Open
Abstract
BACKGROUND Lipolysis is a key metabolic pathway in adipocytes that renders stored triglycerides available for use by other cells and tissues. Non-esterified fatty acids (NEFAs) are known to exert feedback inhibition on adipocyte lipolysis, but the underlying mechanisms have only partly been elucidated. An essential enzyme in adipocyte lipolysis is ATGL. Here, we examined the role of the ATGL inhibitor HILPDA in the negative feedback regulation of adipocyte lipolysis by fatty acids. METHODS We exposed wild-type, HILPDA-deficient and HILPDA-overexpressing adipocytes and mice to various treatments. HILPDA and ATGL protein levels were determined by Western blot. ER stress was assessed by measuring the expression of marker genes and proteins. Lipolysis was studied in vitro and in vivo by measuring NEFA and glycerol levels. RESULTS We show that HILPDA mediates a fatty acid-induced autocrine feedback loop in which elevated intra- or extracellular fatty acids levels upregulate HILPDA by activation of the ER stress response and the fatty acid receptor 4 (FFAR4). The increased HILPDA levels in turn downregulate ATGL protein levels to suppress intracellular lipolysis, thereby maintaining lipid homeostasis. The deficiency of HILPDA under conditions of excessive fatty acid load disrupts this chain of events, leading to elevated lipotoxic stress in adipocytes. CONCLUSION Our data indicate that HILPDA is a lipotoxic marker in adipocytes that mediates a negative feedback regulation of lipolysis by fatty acids via ATGL and alleviates cellular lipotoxic stress.
Collapse
Affiliation(s)
- Lei Deng
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, the Netherlands
| | - Shuangcheng Alivia Wu
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Ling Qi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, 48105, USA; Division of Metabolism, Endocrinology and Diabetes, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, 48105, USA
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, the Netherlands; Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
7
|
Zhao Y, He S, Huang J, Liu M. Genome-Wide Association Analysis of Muscle pH in Texel Sheep × Altay Sheep F 2 Resource Population. Animals (Basel) 2023; 13:2162. [PMID: 37443959 DOI: 10.3390/ani13132162] [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: 05/08/2023] [Revised: 05/29/2023] [Accepted: 06/25/2023] [Indexed: 07/15/2023] Open
Abstract
pH was one of the important meat quality traits, which was an important factor affecting the storage/shelf life and quality of meat in meat production. In order to find a way to extend the storage/shelf life, the pH values (pH45min, pH24h, pH48h and pH72h) of the longissimus dorsi muscles in F2 individuals of 462 Texel sheep × Altay sheep were determined, genotyping was performed using Illumina Ovine SNP 600 K BeadChip and whole genome resequencing technology, a genome-wide association analysis (GWAS) was used to screen the candidate genes and molecular markers for pH values related to the quality traits of mutton, and the effects of population stratification were detected by Q-Q plots. The results showed that the pH population stratification analysis did not find significant systemic bias, and there was no obvious population stratification effect. The results of the association analysis showed that 28 SNPs significantly associated with pH reached the level of genomic significance. The candidate gene associated with pH45min was identified as the CCDC92 gene by gene annotation and a search of the literature. Candidate genes related to pH24h were KDM4C, TGFB2 and GOT2 genes. The candidate genes related to pH48h were MMP12 and MMP13 genes. The candidate genes related to pH72h were HILPDA and FAT1 genes. Further bioinformatics analyses showed 24 gene ontology terms and five signaling pathways that were significantly enriched (p ≤ 0.05). Many terms and pathways were related to cellular components, processes of protein modification, the activity of protein dimerization and hydrolase activity. These identified SNPs and genes could provide useful information about meat and the storage/shelf life of meat, thereby extending the storage/shelf life and quality of meat.
Collapse
Affiliation(s)
- Yilong Zhao
- College of Animal Science and Technology, Shihezi University, Shihezi 832000, China
- College of Animal Science and Technology, Xinjiang Agricultural Vocational and Technical College, Changji 831100, China
| | - Sangang He
- Biotechnology Institute, Xinjiang Academy of Animal Science, Urumqi 830013, China
| | | | - Mingjun Liu
- Biotechnology Institute, Xinjiang Academy of Animal Science, Urumqi 830013, China
| |
Collapse
|
8
|
Adult Triploid Rainbow Trout Can Adapt to Various Dietary Lipid Levels by Coordinating Metabolism in Different Tissues. Metabolites 2023; 13:metabo13030396. [PMID: 36984836 PMCID: PMC10057997 DOI: 10.3390/metabo13030396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Triploid rainbow trout can adapt to various dietary lipid levels; however, the mechanisms of systematic adaptation are not well understood. To investigate how adult triploid rainbow trout maintains lipid hemostasis under different exogenous lipid intake, a 77-day feeding trial was conducted. Diets with lipid contents of 20%, 25%, and 30% were formulated and fed to triploid rainbow trout with an initial weight of 3 ± 0.02 kg, and they were named L20, L25, and L30 group, respectively. Results showed that the condition factor, hepatosomatic index, liver color, and plasma triglyceride were comparable among three groups (p > 0.05), whereas the value of specific growth rate, viscerosomatic index, and liver glycogen content gradually increased with increasing dietary lipid level (p < 0.05). A significantly highest value of plasma glucose and nonesterified fatty acids were found in the L30 group (p < 0.05), whereas the significantly higher content of plasma total cholesterol, high-density lipoprotein–cholesterol, and low-density lipoprotein–cholesterol was found in the L25 group compared with those in L20 group (p < 0.05). As for lipid deposition, abdominal adipose tissue, and muscle were the main lipid storage place for triploid rainbow trout when tissues’ weight is taken into consideration. Overall quantitative PCR showed that the lipid transport and glycolysis were upregulated, and fatty acids oxidative was downregulated in liver when fish were fed low lipid diets. It meant that the liver was the primary lipid metabolizing organ to low lipid diet feeding, which could switch energy supply between glycolysis and fatty acids oxidation. Fish fed with a moderate dietary lipid level diet could increase lipid uptake and promote lipogenesis in muscle. Abdominal adipose tissue could efficiently uptake excess exogenous free fatty acid through upregulating fatty acid uptake and synthesis de novo and then storing it in the form of triglyceride. Excess lipid uptake is preferentially stored in abdominal adipose tissue through coordinated fatty acid uptake and fatty acid synthesis de novo as dietary lipid levels increased. In summary, triploid rainbow trout can adapt to various dietary lipid levels by coordinating metabolism in different tissues.
Collapse
|
9
|
Effect of Leucine-enkephalin on Lipid Deposition and GSK-3β/mTOR Signaling in the Liver of Zebrafish. Int J Pept Res Ther 2023. [DOI: 10.1007/s10989-023-10506-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
|
10
|
Liu L, Liu T, Jia R, Zhang L, Lv Z, He Z, Qu Y, Sun S, Tai F. Downregulation of fatty acid oxidation led by Hilpda increases G2/M arrest/delay-induced kidney fibrosis. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166701. [PMID: 36990128 DOI: 10.1016/j.bbadis.2023.166701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/07/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023]
Abstract
Hypoxia-regulated proximal tubular epithelial cells (PTCs) G2/M phase arrest/delay was involved in production of renal tubulointerstitial fibrosis (TIF). TIF is a common pathological manifestation of progression in patients with chronic kidney disease (CKD), and is often accompanied by lipid accumulation in renal tubules. However, cause-effect relationship between hypoxia-inducible lipid droplet-associated protein (Hilpda), lipid accumulation, G2/M phase arrest/delay and TIF remains unclear. Here we found that overexpression of Hilpda downregulated adipose triglyceride lipase (ATGL) promoted triglyceride overload in the form of lipid accumulation, leading to defective fatty acid β-oxidation (FAO), ATP depletion in a human PTC cell line (HK-2) under hypoxia and in mice kidney tissue treated with unilateral ureteral obstruction (UUO) and unilateral ischemia-reperfusion injury (UIRI). Hilpda-induced lipid accumulation caused mitochondrial dysfunction, enhanced expression of profibrogenic factors TGF-β1, α-SMA and Collagen I elevation, and reduced expression of G2/M phase-associated gene CDK1, as well as increased CyclinB1/D1 ratio, resulted in G2/M phase arrest/delay and profibrogenic phenotypes. Hilpda deficiency in HK-2 cell and kidney of mice with UUO had sustained expression of ATGL and CDK1 and reduced expression of TGF-β1, Collagen I and CyclinB1/D1 ratio, resulting in the amelioration of lipid accumulation and G2/M arrest/delay and subsequent TIF. Expression of Hilpda correlated with lipid accumulation, was positively associated with tubulointerstitial fibrosis in tissue samples from patients with CKD. Our findings suggest that Hilpda deranges fatty acid metabolism in PTCs, which leads to G2/M phase arrest/delay and upregulation of profibrogenic factors, and consequently promote TIF which possibly underlie pathogenesis of CKD.
Collapse
|
11
|
Huang K, Lai S, Guo M, Zhu X, Yuan J, Liu Z, Hu G, Gao Y. Comparison of toxicity between lanthanum oxide nanoparticles and lanthanum chloride. J RARE EARTH 2023. [DOI: 10.1016/j.jre.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
12
|
Gong Z, Li Q, Shi J, Liu ET, Shultz LD, Ren G. Lipid-laden lung mesenchymal cells foster breast cancer metastasis via metabolic reprogramming of tumor cells and natural killer cells. Cell Metab 2022; 34:1960-1976.e9. [PMID: 36476935 PMCID: PMC9819197 DOI: 10.1016/j.cmet.2022.11.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/21/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022]
Abstract
While the distant organ environment is known to support metastasis of primary tumors, its metabolic roles in this process remain underdetermined. Here, in breast cancer models, we found lung-resident mesenchymal cells (MCs) accumulating neutral lipids at the pre-metastatic stage. This was partially mediated by interleukin-1β (IL-1β)-induced hypoxia-inducible lipid droplet-associated (HILPDA) that subsequently represses adipose triglyceride lipase (ATGL) activity in lung MCs. MC-specific ablation of the ATGL or HILPDA genes in mice reinforced and reduced lung metastasis of breast cancer respectively, suggesting a metastasis-promoting effect of lipid-laden MCs. Mechanistically, lipid-laden MCs transported their lipids to tumor cells and natural killer (NK) cells via exosome-like vesicles, leading to heightened tumor cell survival and proliferation and NK cell dysfunction. Blockage of IL-1β, which was effective singly, improved the efficacy of adoptive NK cell immunotherapy in mitigating lung metastasis. Collectively, lung MCs metabolically regulate tumor cells and anti-tumor immunity to facilitate breast cancer lung metastasis.
Collapse
Affiliation(s)
- Zheng Gong
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Qing Li
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Jiayuan Shi
- The Jackson Laboratory, Bar Harbor, ME 04609, USA
| | - Edison T Liu
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | | | - Guangwen Ren
- The Jackson Laboratory, Bar Harbor, ME 04609, USA; Tufts University School of Medicine, Boston, MA 02111, USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
13
|
Zhang R, Meng J, Yang S, Liu W, Shi L, Zeng J, Chang J, Liang B, Liu N, Xing D. Recent Advances on the Role of ATGL in Cancer. Front Oncol 2022; 12:944025. [PMID: 35912266 PMCID: PMC9326118 DOI: 10.3389/fonc.2022.944025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 06/15/2022] [Indexed: 12/22/2022] Open
Abstract
The hypoxic state of the tumor microenvironment leads to reprogramming lipid metabolism in tumor cells. Adipose triglyceride lipase, also known as patatin-like phospholipase= domain-containing protein 2 and Adipose triglyceride lipase (ATGL), as an essential lipid metabolism-regulating enzyme in cells, is regulated accordingly under hypoxia induction. However, studies revealed that ATGL exhibits both tumor-promoting and tumor-suppressing effects, which depend on the cancer cell type and the site of tumorigenesis. For example, elevated ATGL expression in breast cancer is accompanied by enhanced fatty acid oxidation (FAO), enhancing cancer cells’ metastatic ability. In prostate cancer, on the other hand, tumor activity tends to be negatively correlated with ATGL expression. This review outlined the regulation of ATGL-mediated lipid metabolism pathways in tumor cells, emphasizing the Hypoxia-inducible factors 1 (HIF-1)/Hypoxia-inducible lipid droplet-associated (HIG-2)/ATGL axis, peroxisome proliferator-activated receptor (PPAR)/G0/G1 switch gene 2 (G0S2)/ATGL axis, and fat-specific protein 27 (FSP-27)/Early growth response protein 1 (EGR-1)/ATGL axis. In the light of recent research on different cancer types, the role of ATGL on tumorigenesis, tumor proliferation, and tumor metastasis was systemically reviewed.
Collapse
Affiliation(s)
- Renshuai Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Jingsen Meng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Shanbo Yang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Wenjing Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Lingyu Shi
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Jun Zeng
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Jing Chang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Ning Liu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- *Correspondence: Ning Liu, ; Dongming Xing,
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
- School of Life Sciences, Tsinghua University, Beijing, China
- *Correspondence: Ning Liu, ; Dongming Xing,
| |
Collapse
|
14
|
Busato S, Ford HR, Abdelatty AM, Estill CT, Bionaz M. Peroxisome Proliferator-Activated Receptor Activation in Precision-Cut Bovine Liver Slices Reveals Novel Putative PPAR Targets in Periparturient Dairy Cows. Front Vet Sci 2022; 9:931264. [PMID: 35903133 PMCID: PMC9315222 DOI: 10.3389/fvets.2022.931264] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 12/24/2022] Open
Abstract
Metabolic challenges experienced by dairy cows during the transition between pregnancy and lactation (also known as peripartum), are of considerable interest from a nutrigenomic perspective. The mobilization of large amounts of non-esterified fatty acids (NEFA) leads to an increase in NEFA uptake in the liver, the excess of which can cause hepatic accumulation of lipids and ultimately fatty liver. Interestingly, peripartum NEFA activate the Peroxisome Proliferator-activated Receptor (PPAR), a transcriptional regulator with known nutrigenomic properties. The study of PPAR activation in the liver of periparturient dairy cows is thus crucial; however, current in vitro models of the bovine liver are inadequate, and the isolation of primary hepatocytes is time consuming, resource intensive, and prone to errors, with the resulting cells losing characteristic phenotypical traits within hours. The objective of the current study was to evaluate the use of precision-cut liver slices (PCLS) from liver biopsies as a model for PPAR activation in periparturient dairy cows. Three primiparous Jersey cows were enrolled in the experiment, and PCLS from each were prepared prepartum (−8.0 ± 3.6 DIM) and postpartum (+7.7± 1.2 DIM) and treated independently with a variety of PPAR agonists and antagonists: the PPARα agonist WY-14643 and antagonist GW-6471; the PPARδ agonist GW-50156 and antagonist GSK-3787; and the PPARγ agonist rosiglitazone and antagonist GW-9662. Gene expression was assayed through RT-qPCR and RNAseq, and intracellular triacylglycerol (TAG) concentration was measured. PCLS obtained from postpartum cows and treated with a PPARγ agonist displayed upregulation of ACADVL and LIPC while those treated with PPARδ agonist had increased expression of LIPC, PPARD, and PDK4. In PCLS from prepartum cows, transcription of LIPC was increased by all PPAR agonists and NEFA. TAG concentration tended to be larger in tissue slices treated with PPARδ agonist compared to CTR. Use of PPAR isotype-specific antagonists in PCLS cultivated in autologous blood serum failed to decrease expression of PPAR targets, except for PDK4, which was confirmed to be a PPARδ target. Transcriptome sequencing revealed considerable differences in response to PPAR agonists at a false discovery rate-adjusted p-value of 0.2, with the most notable effects exerted by the PPARδ and PPARγ agonists. Differentially expressed genes were mainly related to pathways involved with lipid metabolism and the immune response. Among differentially expressed genes, a subset of 91 genes were identified as novel putative PPAR targets in the bovine liver, by cross-referencing our results with a publicly available dataset of predicted PPAR target genes, and supplementing our findings with prior literature. Our results provide important insights on the use of PCLS as a model for assaying PPAR activation in the periparturient dairy cow.
Collapse
Affiliation(s)
- Sebastiano Busato
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Hunter R. Ford
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
| | - Alzahraa M. Abdelatty
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Charles T. Estill
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
- College of Veterinary Medicine, Oregon State University, Corvallis, OR, United States
| | - Massimo Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis, OR, United States
- *Correspondence: Massimo Bionaz
| |
Collapse
|
15
|
Erol SA, Anuk AT, Tanaçan A, Semiz H, Keskin HL, Neşelioğlu S, Erel Ö, Moraloğlu Tekin Ö, Şahin D. An evaluation of maternal serum dynamic thiol-disulfide homeostasis and ischemia modified albumin changes in pregnant women with COVID-19. Turk J Obstet Gynecol 2022; 19:21-27. [PMID: 35343216 PMCID: PMC8966320 DOI: 10.4274/tjod.galenos.2022.72929] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: It is thought that oxidative stress, free radicals, reactive oxygen species and reactive nitrogen species affect the pathophysiology of coronavirus disease-2019 (COVID-19). This study aimed to evaluate the oxidative status in pregnant patients with COVID-19 infection according to the changes seen in the levels of maternal serum thiol-disulfide and ischemia-modified albumin (IMA). Materials and Methods: A study group was formed of 40 pregnant women with confirmed COVID-19 infection (study group) and a control group of 40 healthy pregnant women with no risk factors determined. In this prospective, case-controlled study, analyses were made of the maternal serum native thiol, total thiol, disulfide, IMA, and disulfide/native thiol concentrations. Results: The maternal serum native thiol and total thiol concentrations in the study group were determined to be statistically significantly lower (p=0.007 and p=0.006, respectively), and the disulfide/native thiol ratio was higher but not to a level of statistical significance (p=0.473). There was no difference between the two groups regarding IMA levels (p=0.731). Conclusion: The thiol-disulfide balance was seen to shift in the oxidant direction in pregnancies with COVID-19, which might support the view that ischemic processes play a role in the etiopathogenesis of this novel disease.
Collapse
|
16
|
Triglyceride breakdown from lipid droplets regulates the inflammatory response in macrophages. Proc Natl Acad Sci U S A 2022; 119:e2114739119. [PMID: 35302892 PMCID: PMC8944848 DOI: 10.1073/pnas.2114739119] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lipid droplets (LDs) are ubiquitous organelles that play important roles in cellular energy homeostasis, tightly regulating the accumulation and release of lipids. In macrophages, lipids accumulate in LDs during inflammation. However, it is unclear how inflammatory activation promotes the accumulation of lipids in LDs, and how the dynamic between lipid accumulation and breakdown could drive or inhibit inflammation. Elucidating the role of lipid accumulation during inflammation may provide important knowledge to influence inflammatory processes during health and disease. We identify the importance of the hypoxia-inducible lipid droplet–associated protein and the intracellular adipose triglyceride lipase in the regulation of lipid accumulation and breakdown in inflammatory macrophages. Furthermore, we determine the regulatory effect of lipid breakdown from LDs in supporting inflammation. In response to inflammatory activation by pathogens, macrophages accumulate triglycerides in intracellular lipid droplets. The mechanisms underlying triglyceride accumulation and its exact role in the inflammatory response of macrophages are not fully understood. Here, we aim to further elucidate the mechanism and function of triglyceride accumulation in the inflammatory response of activated macrophages. Lipopolysaccharide (LPS)-mediated activation markedly increased triglyceride accumulation in macrophages. This increase could be attributed to up-regulation of the hypoxia-inducible lipid droplet–associated (HILPDA) protein, which down-regulated adipose triglyceride lipase (ATGL) protein levels, in turn leading to decreased ATGL-mediated triglyceride hydrolysis. The reduction in ATGL-mediated lipolysis attenuated the inflammatory response in macrophages after ex vivo and in vitro activation, and was accompanied by decreased production of prostaglandin-E2 (PGE2) and interleukin-6 (IL-6). Overall, we provide evidence that LPS-mediated activation of macrophages suppresses lipolysis via induction of HILPDA, thereby reducing the availability of proinflammatory lipid precursors and suppressing the production of PGE2 and IL-6.
Collapse
|
17
|
Mice with a deficiency in Peroxisomal Membrane Protein 4 (PXMP4) display mild changes in hepatic lipid metabolism. Sci Rep 2022; 12:2512. [PMID: 35169201 PMCID: PMC8847483 DOI: 10.1038/s41598-022-06479-y] [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: 10/28/2020] [Accepted: 01/31/2022] [Indexed: 11/08/2022] Open
Abstract
Peroxisomes play an important role in the metabolism of a variety of biomolecules, including lipids and bile acids. Peroxisomal Membrane Protein 4 (PXMP4) is a ubiquitously expressed peroxisomal membrane protein that is transcriptionally regulated by peroxisome proliferator-activated receptor α (PPARα), but its function is still unknown. To investigate the physiological function of PXMP4, we generated a Pxmp4 knockout (Pxmp4-/-) mouse model using CRISPR/Cas9-mediated gene editing. Peroxisome function was studied under standard chow-fed conditions and after stimulation of peroxisomal activity using the PPARα ligand fenofibrate or by using phytol, a metabolite of chlorophyll that undergoes peroxisomal oxidation. Pxmp4-/- mice were viable, fertile, and displayed no changes in peroxisome numbers or morphology under standard conditions. Also, no differences were observed in the plasma levels of products from major peroxisomal pathways, including very long-chain fatty acids (VLCFAs), bile acids (BAs), and BA intermediates di- and trihydroxycholestanoic acid. Although elevated levels of the phytol metabolites phytanic and pristanic acid in Pxmp4-/- mice pointed towards an impairment in peroxisomal α-oxidation capacity, treatment of Pxmp4-/- mice with a phytol-enriched diet did not further increase phytanic/pristanic acid levels. Finally, lipidomic analysis revealed that loss of Pxmp4 decreased hepatic levels of the alkyldiacylglycerol class of neutral ether lipids, particularly those containing polyunsaturated fatty acids. Together, our data show that while PXMP4 is not critical for overall peroxisome function under the conditions tested, it may have a role in the metabolism of (ether)lipids.
Collapse
|
18
|
Guo J, Zhang M, Wang H, Li N, Lu Z, Li L, Hui S, Xu H. Gut microbiota and short chain fatty acids partially mediate the beneficial effects of inulin on metabolic disorders in obese ob/ob mice. J Food Biochem 2022; 46:e14063. [PMID: 35128673 DOI: 10.1111/jfbc.14063] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/25/2021] [Accepted: 12/06/2021] [Indexed: 02/01/2023]
Abstract
Mounting evidence has linked both obesity and metabolic disorders with dysbiosis of the gut microbiota. Dietary inulin is conducive to modulating this dysbiosis, and represents a potential means to improve disorders of glucose and lipid metabolism. However, the mechanisms underlying these improvements are largely unclear. Obese ob/ob mice were fed a standard chow, a low fiber diet (LFD) or a high fiber diet (HFD) for 4 weeks, and the body weight, fecal short chain fatty acids (SCFAs) level, and plasma and liver lipid profiles were analyzed. Oral glucose tolerance testing, and gut microbiota sequencing were also conducted. Dietary inulin improved the dysbiosis of the gut microbiota, attenuated the decrease in phylum Bacteroidetes, repressed the increase of phylum Firmicutes, and led to an increase in the ratio of Firmicutes/Bacteroidetes. At the family level, inulin promoted the expansion of SCFAs-producing Ruminococcaceae and Lachnospiraceae bacteria, which increased the fecal SCFAs concentrations. At the genus level, inulin increased the levels of Bacteroides and Bifidobacteria. Furthermore, our results revealed that there was enhanced expression of angiopoietin-like protein 4 (ANGPTL4), which might be induced by the higher production of SCFAs, and this may underlie the improvements in the disorders of glucose and lipid metabolism seen in mice with added dietary inulin. In conclusion, inulin may ameliorate metabolic disorders by remodeling the gut microbiota and increasing the production of SCFAs, which might be mediated by the ANGPTL4-related signaling pathway. Interventions targeting the gut microbiota warrant further investigation as a novel therapy for metabolic diseases. PRACTICAL APPLICATIONS: Mounting evidence has linked both obesity and metabolic disorders with dysbiosis of the gut microbiota. Dietary inulin is conducive to modulating this dysbiosis, and represents a potential means to improve disorders of glucose and lipid metabolism. However, the mechanisms underlying these improvements are largely unclear. In the present study, we investigated the effects of dietary fiber (inulin) on metabolic homeostasis using ob/ob mice. The results of our study demonstrate that inulin-induced remodeling of the gut microbiota resulted in increased production of short chain fatty acids (SCFAs), leading to the enhanced expression of angiopoietin-like protein 4 (ANGPTL4), which improved the glucose and lipid metabolism. Our results suggest that the gut microbiota, SCFAs and ANGPTL4 pathway at least partially mediate the beneficial effects of inulin on metabolic disorders in ob/ob mice.
Collapse
Affiliation(s)
- Jing Guo
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mengyuan Zhang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - He Wang
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Na Li
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zongliang Lu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Long Li
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Suocheng Hui
- Research Department, The Rocket Force Characteristic Medical Center of Chinese People's Liberation Army, Beijing, China
| | - Hongxia Xu
- Department of Clinical Nutrition, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
19
|
Adipose Triglyceride Lipase in Hepatic Physiology and Pathophysiology. Biomolecules 2021; 12:biom12010057. [PMID: 35053204 PMCID: PMC8773762 DOI: 10.3390/biom12010057] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/28/2021] [Accepted: 12/28/2021] [Indexed: 12/25/2022] Open
Abstract
The liver is extremely active in oxidizing triglycerides (TG) for energy production. An imbalance between TG synthesis and hydrolysis leads to metabolic disorders in the liver, including excessive lipid accumulation, oxidative stress, and ultimately liver damage. Adipose triglyceride lipase (ATGL) is the rate-limiting enzyme that catalyzes the first step of TG breakdown to glycerol and fatty acids. Although its role in controlling lipid homeostasis has been relatively well-studied in the adipose tissue, heart, and skeletal muscle, it remains largely unknown how and to what extent ATGL is regulated in the liver, responds to stimuli and regulators, and mediates disease progression. Therefore, in this review, we describe the current understanding of the structure–function relationship of ATGL, the molecular mechanisms of ATGL regulation at translational and post-translational levels, and—most importantly—its role in lipid and glucose homeostasis in health and disease with a focus on the liver. Advances in understanding the molecular mechanisms underlying hepatic lipid accumulation are crucial to the development of targeted therapies for treating hepatic metabolic disorders.
Collapse
|
20
|
Liu D, Yu H, Gu Y, Pang Q. Effect of rare earth element lanthanum on lipid deposition and Wnt10b signaling in the liver of male zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 240:105994. [PMID: 34656894 DOI: 10.1016/j.aquatox.2021.105994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 06/13/2023]
Abstract
This paper investigates the effect of lanthanum (La) on lipid deposition and Wnt10b signaling in the liver of male zebrafish with exposure of 0, 10, 20, and 30 μmol/L La. It suggests that La can be accumulated in liver, and its treatments decrease the activities and gene expression of enzymes related to fatty acid synthesis. The levels of total cholesterol (TC), triglyceride (TG), and nonesterified fatty acids (NEFA) as well as the size of lipid droplets are decreased by La treatments. Moreover, La treatments affect the composition of fatty acids and the content of nutrient elements. Meanwhile, they also induce the gene expression of wnt10b, β-catenin, pparα, and pparγ, but inhibit gsk-3β gene expression in liver. Further study on the result of wnt10b gene interference shows that Wnt10b/β-catenin signaling plays a crucial role in the regulatory process of hepatic lipid deposition. Taken together, our observations suggest that La accumulation affects lipid deposition in the liver of male zebrafish, and Wnt10b signaling pathway may be involved in this process.
Collapse
Affiliation(s)
- Dongwu Liu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo 255049, China; Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China.
| | - Hairui Yu
- College of Biological and Agricultural Engineering, Weifang Bioengineering Technology Research Center, Weifang University, Weifang 261061, China
| | - Yaqi Gu
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China
| | - Qiuxiang Pang
- Anti-aging & Regenerative Medicine Research Institution, School of Life Sciences, Shandong University of Technology, Zibo 255049, China.
| |
Collapse
|
21
|
Grabner GF, Xie H, Schweiger M, Zechner R. Lipolysis: cellular mechanisms for lipid mobilization from fat stores. Nat Metab 2021; 3:1445-1465. [PMID: 34799702 DOI: 10.1038/s42255-021-00493-6] [Citation(s) in RCA: 364] [Impact Index Per Article: 91.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/15/2021] [Indexed: 12/13/2022]
Abstract
The perception that intracellular lipolysis is a straightforward process that releases fatty acids from fat stores in adipose tissue to generate energy has experienced major revisions over the last two decades. The discovery of new lipolytic enzymes and coregulators, the demonstration that lipophagy and lysosomal lipolysis contribute to the degradation of cellular lipid stores and the characterization of numerous factors and signalling pathways that regulate lipid hydrolysis on transcriptional and post-transcriptional levels have revolutionized our understanding of lipolysis. In this review, we focus on the mechanisms that facilitate intracellular fatty-acid mobilization, drawing on canonical and noncanonical enzymatic pathways. We summarize how intracellular lipolysis affects lipid-mediated signalling, metabolic regulation and energy homeostasis in multiple organs. Finally, we examine how these processes affect pathogenesis and how lipolysis may be targeted to potentially prevent or treat various diseases.
Collapse
Affiliation(s)
- Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Hao Xie
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Martina Schweiger
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, Graz, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
22
|
Schiffrin M, Winkler C, Quignodon L, Naldi A, Trötzmüller M, Köfeler H, Henry H, Parini P, Desvergne B, Gilardi F. Sex Dimorphism of Nonalcoholic Fatty Liver Disease (NAFLD) in Pparg-Null Mice. Int J Mol Sci 2021; 22:9969. [PMID: 34576136 PMCID: PMC8467431 DOI: 10.3390/ijms22189969] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/09/2021] [Accepted: 09/11/2021] [Indexed: 12/12/2022] Open
Abstract
Men with nonalcoholic fatty liver disease (NAFLD) are more exposed to nonalcoholic steatohepatitis (NASH) and liver fibrosis than women. However, the underlying molecular mechanisms of NALFD sex dimorphism are unclear. We combined gene expression, histological and lipidomic analyses to systematically compare male and female liver steatosis. We characterized hepatosteatosis in three independent mouse models of NAFLD, ob/ob and lipodystrophic fat-specific (PpargFΔ/Δ) and whole-body PPARγ-null (PpargΔ/Δ) mice. We identified a clear sex dimorphism occurring only in PpargΔ/Δ mice, with females showing macro- and microvesicular hepatosteatosis throughout their entire life, while males had fewer lipid droplets starting from 20 weeks. This sex dimorphism in hepatosteatosis was lost in gonadectomized PpargΔ/Δ mice. Lipidomics revealed hepatic accumulation of short and highly saturated TGs in females, while TGs were enriched in long and unsaturated hydrocarbon chains in males. Strikingly, sex-biased genes were particularly perturbed in both sexes, affecting lipid metabolism, drug metabolism, inflammatory and cellular stress response pathways. Most importantly, we found that the expression of key sex-biased genes was severely affected in all the NAFLD models we tested. Thus, hepatosteatosis strongly affects hepatic sex-biased gene expression. With NAFLD increasing in prevalence, this emphasizes the urgent need to specifically address the consequences of this deregulation in humans.
Collapse
Affiliation(s)
- Mariano Schiffrin
- Center of Integrative Genomics, Genopode, Lausanne Faculty of Biology and Medicine, CH-1015 Lausanne, Switzerland; (M.S.); (C.W.); (L.Q.); (A.N.); (B.D.)
| | - Carine Winkler
- Center of Integrative Genomics, Genopode, Lausanne Faculty of Biology and Medicine, CH-1015 Lausanne, Switzerland; (M.S.); (C.W.); (L.Q.); (A.N.); (B.D.)
| | - Laure Quignodon
- Center of Integrative Genomics, Genopode, Lausanne Faculty of Biology and Medicine, CH-1015 Lausanne, Switzerland; (M.S.); (C.W.); (L.Q.); (A.N.); (B.D.)
| | - Aurélien Naldi
- Center of Integrative Genomics, Genopode, Lausanne Faculty of Biology and Medicine, CH-1015 Lausanne, Switzerland; (M.S.); (C.W.); (L.Q.); (A.N.); (B.D.)
| | - Martin Trötzmüller
- Core Facility Mass Spectrometry, Medical University of Graz, 8036 Graz, Austria; (M.T.); (H.K.)
| | - Harald Köfeler
- Core Facility Mass Spectrometry, Medical University of Graz, 8036 Graz, Austria; (M.T.); (H.K.)
| | - Hugues Henry
- Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne Faculty of Biology and Medicine, CH-1011 Lausanne, Switzerland;
| | - Paolo Parini
- CardioMetabolic Unit, Department of Medicine and Department of Laboratory Medicine, Karolinska Insititutet and Theme Inflammation and Ageing Karolinska University Hospital Huddinge, 14186 Stockholm, Sweden;
| | - Béatrice Desvergne
- Center of Integrative Genomics, Genopode, Lausanne Faculty of Biology and Medicine, CH-1015 Lausanne, Switzerland; (M.S.); (C.W.); (L.Q.); (A.N.); (B.D.)
| | - Federica Gilardi
- Center of Integrative Genomics, Genopode, Lausanne Faculty of Biology and Medicine, CH-1015 Lausanne, Switzerland; (M.S.); (C.W.); (L.Q.); (A.N.); (B.D.)
- Faculty Unit of Toxicology, University Center of Legal Medicine, Faculty of Biology and Medicine, Lausanne University Hospital, CH-1000 Lausanne, Switzerland
| |
Collapse
|
23
|
Defour M, van Weeghel M, Hermans J, Kersten S. Hepatic ADTRP overexpression does not influence lipid and glucose metabolism. Am J Physiol Cell Physiol 2021; 321:C585-C595. [PMID: 34288722 DOI: 10.1152/ajpcell.00185.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The peroxisome proliferator activated receptors (PPARs) are a group of transcription factors belonging to the nuclear receptor superfamily. Since most target genes of either PPARs are implicated in lipid and glucose metabolism, regulation by PPARs could be used as a screening tool to identify novel genes involved in lipid or glucose metabolism. Here, we identify Adtrp, a serine hydrolase enzyme that was reported to catalyze the hydrolysis of fatty acid esters of hydroxy fatty acids (FAHFAs), as a novel PPAR-regulated gene. Adtrp was significantly upregulated by PPARα activation in mouse primary hepatocytes, liver slices, and whole liver. In addition, Adtrp was upregulated by PPARγ activation in 3L3-L1 adipocytes and in white adipose tissue. ChIP-SEQ identified a strong PPAR binding site in the immediate upstream promoter of the Adtrp gene. Adenoviral-mediated hepatic overexpression of Adtrp in diet-induced obese mice caused a modest increase in plasma non-esterified fatty acids but did not influence diet-induced obesity, liver triglyceride levels, liver lipidomic profiles, liver transcriptomic profiles, and plasma cholesterol, triglycerides, glycerol, and glucose levels. Moreover, hepatic Adtrp overexpression did not lead to significant changes in FAHFA levels in plasma or liver and did not influence glucose and insulin tolerance. Finally, hepatic overexpression of Adtrp did not influence liver triglycerides and levels of plasma metabolites after a 24h fast. Taken together, our data suggest that despite being a PPAR-regulated gene, hepatic Adtrp does not seem to play a major role in lipid and glucose metabolism and does not regulate FAHFA levels.
Collapse
Affiliation(s)
- Merel Defour
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, Wageningen, Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Meibergdreef Amsterdam, Amsterdam, Netherlands
| | - Jill Hermans
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam, The Netherlands; Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Meibergdreef Amsterdam, Amsterdam, Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, Wageningen, Netherlands
| |
Collapse
|
24
|
Shakya S, Gromovsky AD, Hale JS, Knudsen AM, Prager B, Wallace LC, Penalva LOF, Brown HA, Kristensen BW, Rich JN, Lathia JD, Brown JM, Hubert CG. Altered lipid metabolism marks glioblastoma stem and non-stem cells in separate tumor niches. Acta Neuropathol Commun 2021; 9:101. [PMID: 34059134 PMCID: PMC8166002 DOI: 10.1186/s40478-021-01205-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 05/19/2021] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma (GBM) displays marked cellular and metabolic heterogeneity that varies among cellular microenvironments within a tumor. Metabolic targeting has long been advocated as a therapy against many tumors including GBM, but how lipid metabolism is altered to suit different microenvironmental conditions and whether cancer stem cells (CSCs) have altered lipid metabolism are outstanding questions in the field. We interrogated gene expression in separate microenvironments of GBM organoid models that mimic the transition between nutrient-rich and nutrient-poor pseudopalisading/perinecrotic tumor zones using spatial-capture RNA-sequencing. We revealed a striking difference in lipid processing gene expression and total lipid content between diverse cell populations from the same patient, with lipid enrichment in hypoxic organoid cores and also in perinecrotic and pseudopalisading regions of primary patient tumors. This was accompanied by regionally restricted upregulation of hypoxia-inducible lipid droplet-associated (HILPDA) gene expression in organoid cores and pseudopalisading regions of clinical GBM specimens, but not lower-grade brain tumors. CSCs have low lipid droplet accumulation compared to non-CSCs in organoid models and xenograft tumors, and prospectively sorted lipid-low GBM cells are functionally enriched for stem cell activity. Targeted lipidomic analysis of multiple patient-derived models revealed a significant shift in lipid metabolism between GBM CSCs and non-CSCs, suggesting that lipid levels may not be simply a product of the microenvironment but also may be a reflection of cellular state. CSCs had decreased levels of major classes of neutral lipids compared to non-CSCs, but had significantly increased polyunsaturated fatty acid production due to high fatty acid desaturase (FADS1/2) expression which was essential to maintain CSC viability and self-renewal. Our data demonstrate spatially and hierarchically distinct lipid metabolism phenotypes occur clinically in the majority of patients, can be recapitulated in laboratory models, and may represent therapeutic targets for GBM.
Collapse
Affiliation(s)
- Sajina Shakya
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH ND2-4044195 USA
| | - Anthony D. Gromovsky
- Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH USA
| | - James S. Hale
- Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH USA
| | - Arnon M. Knudsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Briana Prager
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH USA
- Medical Scientist Training Program, Case Western Reserve School of Medicine, Cleveland, OH USA
| | - Lisa C. Wallace
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH ND2-4044195 USA
| | - Luiz O. F. Penalva
- Children’s Cancer Research Institute, University of Texas Health Sciences Center San Antonio, San Antonio, TX USA
| | - H. Alex Brown
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN USA
| | - Bjarne W. Kristensen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jeremy N. Rich
- Department of Medicine, University of Pittsburgh, Pittsburg, PA USA
| | - Justin D. Lathia
- Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH USA
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH USA
| | - J. Mark Brown
- Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH USA
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH USA
| | - Christopher G. Hubert
- Department of Biomedical Engineering, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH ND2-4044195 USA
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH USA
- Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, Cleveland, OH USA
| |
Collapse
|
25
|
Grachan JJ, Kery M, Giaccia AJ, Denko NC, Papandreou I. Lipid droplet storage promotes murine pancreatic tumor growth. Oncol Rep 2021; 45:21. [PMID: 33649859 PMCID: PMC8889526 DOI: 10.3892/or.2021.7972] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/20/2020] [Indexed: 11/06/2022] Open
Abstract
Hypoxia Inducible Lipid Droplet Associated (HILPDA) is frequently overexpressed in tumors and promotes neutral lipid storage. The impact of Hilpda on pancreatic ductal adenocarcinoma (PDAC) tumor growth is not known. In order to evaluate Hilpda‑dependent lipid storage mechanisms, expression of Hilpda in murine pancreatic cells (KPC) was genetically manipulated. Lipid droplet (LD) abundance and triglyceride content in vitro were measured, and model tumor growth in nu/nu mice was determined. The results showed that excess lipid supply increased triglyceride storage and LD formation in KPC cells in a HILPDA‑dependent manner. Contrary to published results, inhibition of Adipose Triglyceride Lipase (ATGL) did not ameliorate the triglyceride abundance differences between Hilpda WT and KO cells. Hilpda ablation significantly decreased the growth rate of model tumors in immunocompromised mice. In conclusion, Hilpda is a positive regulator of triglyceride storage and lipid droplet formation in murine pancreatic cancer cells in vitro and lipid accumulation and tumor growth in vivo. Our data suggest that deregulated ATGL is not responsible for the absence of LDs in KO cells in this context.
Collapse
Affiliation(s)
- Jeremy J. Grachan
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Martin Kery
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Nicholas C. Denko
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Ioanna Papandreou
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
- Correspondence to: Dr Ioanna Papandreou, Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 420 W. 12th Avenue, Columbus, OH 43210, USA, E-mail:
| |
Collapse
|
26
|
van Dierendonck XAMH, de la Rosa Rodriguez MA, Georgiadi A, Mattijssen F, Dijk W, van Weeghel M, Singh R, Borst JW, Stienstra R, Kersten S. HILPDA Uncouples Lipid Droplet Accumulation in Adipose Tissue Macrophages from Inflammation and Metabolic Dysregulation. Cell Rep 2021; 30:1811-1822.e6. [PMID: 32049012 DOI: 10.1016/j.celrep.2020.01.046] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 12/10/2019] [Accepted: 01/15/2020] [Indexed: 01/15/2023] Open
Abstract
Obesity leads to a state of chronic, low-grade inflammation that features the accumulation of lipid-laden macrophages in adipose tissue. Here, we determined the role of macrophage lipid-droplet accumulation in the development of obesity-induced adipose-tissue inflammation, using mice with myeloid-specific deficiency of the lipid-inducible HILPDA protein. HILPDA deficiency markedly reduced intracellular lipid levels and accumulation of fluorescently labeled fatty acids. Decreased lipid storage in HILPDA-deficient macrophages can be rescued by inhibition of adipose triglyceride lipase (ATGL) and is associated with increased oxidative metabolism. In diet-induced obese mice, HILPDA deficiency does not alter inflammatory and metabolic parameters, despite markedly reducing lipid accumulation in macrophages. Overall, we find that HILPDA is a lipid-inducible, physiological inhibitor of ATGL-mediated lipolysis in macrophages and uncouples lipid storage in adipose tissue macrophages from inflammation and metabolic dysregulation. Our data question the contribution of lipid droplet accumulation in adipose tissue macrophages in obesity-induced inflammation and metabolic dysregulation.
Collapse
Affiliation(s)
- Xanthe A M H van Dierendonck
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein 8, 6525 GA Nijmegen, the Netherlands
| | - Montserrat A de la Rosa Rodriguez
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Anastasia Georgiadi
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Frits Mattijssen
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Wieneke Dijk
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands; Gastroenterology and Metabolism, Amsterdam Cardiovascular Sciences, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Rajat Singh
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 505D, Bronx, NY 10461, USA
| | - Jan Willem Borst
- Laboratory of Biochemistry, Microspectroscopy Research Facility, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands
| | - Rinke Stienstra
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Geert Grooteplein 8, 6525 GA Nijmegen, the Netherlands.
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE Wageningen, the Netherlands.
| |
Collapse
|
27
|
de la Rosa Rodriguez MA, Deng L, Gemmink A, van Weeghel M, Aoun ML, Warnecke C, Singh R, Borst JW, Kersten S. Hypoxia-inducible lipid droplet-associated induces DGAT1 and promotes lipid storage in hepatocytes. Mol Metab 2021; 47:101168. [PMID: 33465519 PMCID: PMC7881268 DOI: 10.1016/j.molmet.2021.101168] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 01/08/2021] [Accepted: 01/12/2021] [Indexed: 12/26/2022] Open
Abstract
Objective Storage of triglycerides in lipid droplets is governed by a set of lipid droplet-associated proteins. One of these lipid droplet-associated proteins, hypoxia-inducible lipid droplet-associated (HILPDA), was found to impair lipid droplet breakdown in macrophages and cancer cells by inhibiting adipose triglyceride lipase. Here, we aimed to better characterize the role and mechanism of action of HILPDA in hepatocytes. Methods We performed studies in HILPDA-deficient and HILPDA-overexpressing liver cells, liver slices, and mice. The functional role and physical interactions of HILPDA were investigated using a variety of biochemical and microscopic techniques, including real-time fluorescence live-cell imaging and Förster resonance energy transfer-fluorescence lifetime imaging microscopy (FRET-FLIM). Results Levels of HILPDA were markedly induced by fatty acids in several hepatoma cell lines. Hepatocyte-specific deficiency of HILPDA in mice modestly but significantly reduced hepatic triglycerides in mice with non-alcoholic steatohepatitis. Similarly, deficiency of HILPDA in mouse liver slices and primary hepatocytes reduced lipid storage and accumulation of fluorescently-labeled fatty acids in lipid droplets, respectively, which was independent of adipose triglyceride lipase. Fluorescence microscopy showed that HILPDA partly colocalizes with lipid droplets and with the endoplasmic reticulum, is especially abundant in perinuclear areas, and mainly associates with newly added fatty acids. Real-time fluorescence live-cell imaging further revealed that HILPDA preferentially localizes to lipid droplets that are being remodeled. Overexpression of HILPDA in liver cells increased the activity of diacylglycerol acyltransferases (DGAT) and DGAT1 protein levels, concurrent with increased lipid storage. Confocal microscopy coupled to FRET-FLIM analysis demonstrated that HILPDA physically interacts with DGAT1 in living liver cells. The stimulatory effect of HILPDA on lipid storage via DGAT1 was corroborated in adipocytes. Conclusions Our data indicate that HILPDA physically interacts with DGAT1 and increases DGAT activity. Our findings suggest a novel regulatory mechanism by which fatty acids promote triglyceride synthesis and storage. HILPDA expression is induced by fatty acids in hepatoma cells. HILPDA deficiency modestly decreases liver triglyceride storage in mice with NASH. HILPDA preferentially associates with newly synthesized lipid droplets and active lipid droplets. HILPDA promotes lipid storage at least in part independently of ATGL. HILPDA physically interacts and induces DGAT1.
Collapse
Affiliation(s)
- Montserrat A de la Rosa Rodriguez
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| | - Lei Deng
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| | - Anne Gemmink
- Department of Nutrition and Movement Sciences, Maastricht University Medical Center+, Maastricht, 6200 MD, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, the Netherlands
| | - Marie Louise Aoun
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 505D, Bronx, NY, 10461, USA
| | - Christina Warnecke
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Rajat Singh
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Forchheimer 505D, Bronx, NY, 10461, USA
| | - Jan Willem Borst
- Laboratory of Biochemistry, Microspectroscopy Research Facility, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, Wageningen, 6708 WE, the Netherlands.
| |
Collapse
|
28
|
Povero D, Johnson SM, Liu J. Hypoxia, hypoxia-inducible gene 2 (HIG2)/HILPDA, and intracellular lipolysis in cancer. Cancer Lett 2020; 493:71-79. [PMID: 32818550 PMCID: PMC11218043 DOI: 10.1016/j.canlet.2020.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/27/2020] [Accepted: 06/13/2020] [Indexed: 12/16/2022]
Abstract
Tumor tissues are chronically exposed to hypoxia owing to aberrant vascularity. Hypoxia induces metabolic alterations in cancer, thereby promoting aggressive malignancy and metastasis. While previous efforts largely focused on adaptive responses in glucose and glutamine metabolism, recent studies have begun to yield important insight into the hypoxic regulation of lipid metabolic reprogramming in cancer. Emerging evidence points to lipid droplet (LD) accumulation as a hallmark of hypoxic cancer cells. One critical underlying mechanism involves the inhibition of adipose triglyceride lipase (ATGL)-mediated intracellular lipolysis by a small protein encoded by hypoxia-inducible gene 2 (HIG2), also known as hypoxia inducible lipid droplet associated (HILPDA). In this review we summarize and discuss recent key findings on hypoxia-dependent regulation of metabolic adaptations especially lipolysis in cancer. We also pose several questions and hypotheses pertaining to the metabolic impact of lipolytic regulation in cancer under hypoxia and during hypoxia-reoxygenation transition.
Collapse
Affiliation(s)
- Davide Povero
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA
| | - Scott M Johnson
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Mayo Clinic College of Medicine & Science, Rochester, MN, 55905, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, 55905, USA
| | - Jun Liu
- From Department of Biochemistry and Molecular Biology, Rochester, MN, 55905, USA; Division of Endocrinology, Rochester, MN, 55905, USA.
| |
Collapse
|
29
|
Xiang W, Cheng S, Zhou Y, Ma L. Effects of 1,25(OH) 2 D 3 on lipid droplet growth in adipocytes. Biofactors 2020; 46:943-954. [PMID: 31904171 DOI: 10.1002/biof.1610] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 12/22/2019] [Indexed: 02/06/2023]
Abstract
This study aimed to explore the effects of 1,25(OH)2 D3 on lipid droplet (LD) growth in 3T3-L1 adipocytes of hypertrophy model. Cocktail method was used to induce differentiation in 3T3-L1 cells. After 8 days, the cells were modeled by 100, 300, 600, and 900 μM palmitic acid (PA) for 24 hr. The best concentration of modeling was screened by MTT results and triglycerides (TG) content. The model cells were intervened by 1, 10, and 100 nM 1,25(OH)2 D3 for 24 hr. Then, the TG content of cells were detected and stained by oil red O. The diameter and quantity of LDs were analyzed. mRNA relative expression levels of genes related to LD (CIDE-a, Fsp27, PLIN-1), upstream response factor (PPAR-α, PPAR-γ, and VDR), and TG metabolism (long chain acyl-CoA synthetase 3, 1-acylglycerol-3-phosphate O-acyltransferase 1, adipose triglyceride lipase, diacylglycerol acyltransferase 1, diacylglycerol acyltransferase 2, glycerol-3-phosphate O-acyltransferase 3, glycerol-3-phosphate O-acyltransferase 4, hormone-sensitive lipase, mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-acetyl glucosaminyl transferase, phosphatidic acid phosphatase, and uncoupling protein-1) were detected by RT-qPCR. A total of 300 μM PA was selected as the optimum concentration. Compared with model group, 10 and 100 nM 1,25(OH)2 D3 decreased the average diameter, increased the quantity of LDs, upregulated PPAR-α and PLIN-1 mRNA expression levels, and downregulated CIDE-a and Fsp27 mRNA expression levels significantly (p < .05). However, 1 nM 1,25(OH)2 D3 did not alter LD morphology and TG content. mRNA expression levels of long chain acyl-CoA synthetase 3, 1-acylglycerol-3-phosphate O-acyltransferase 1, diacylglycerol acyltransferase 2, glycerol-3-phosphate O-acyltransferase 3, and glycerol-3-phosphate O-acyltransferase 4 in 10 and 100 nM groups were significantly lower than those in the model group (p < .05); mRNA expression levels of adipose triglyceride lipase, diacylglycerol acyltransferase 1, hormone-sensitive lipase, mannosyl (alpha-1,3-)-glycoprotein beta-1,2-N-acetyl glucosaminyl transferase, phosphatidic acid phosphatase, and uncoupling protein-1 were significantly increased in the 100 nM group (p < .05). The 10 and 100 nM 1,25(OH)2 D3 can inhibit LD fusion, promote LD decomposition, reduce LD volume, and inhibit lipogenesis through the PPAR-α signaling pathway.
Collapse
Affiliation(s)
- Wei Xiang
- Department of Nutrition and Diet, Changzhou Traditional Chinese Medicine Hospital, Changzhou, China
| | - Shi Cheng
- Department of Nutrition and Food Hygiene, School of Public Health, Xinjiang Medical University, Urumqi, China
| | - Yong Zhou
- Department of Medical Cell Biology and Genetics, College of Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Ling Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Southwest Medical University, Luzhou, China
| |
Collapse
|
30
|
de la Rosa Rodriguez MA, Kersten S. Regulation of lipid droplet homeostasis by hypoxia inducible lipid droplet associated HILPDA. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158738. [PMID: 32417386 DOI: 10.1016/j.bbalip.2020.158738] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/17/2020] [Accepted: 05/06/2020] [Indexed: 12/28/2022]
Abstract
Nearly all cell types have the ability to store excess energy as triglycerides in specialized organelles called lipid droplets. The formation and degradation of lipid droplets is governed by a diverse set of enzymes and lipid droplet-associated proteins. One of the lipid droplet-associated proteins is Hypoxia Inducible Lipid Droplet Associated (HILPDA). HILPDA was originally discovered in a screen to identify novel hypoxia-inducible proteins. Apart from hypoxia, levels of HILPDA are induced by fatty acids and adrenergic agonists. HILPDA is a small protein of 63 amino acids in humans and 64 amino acids in mice. Inside cells, HILPDA is located in the endoplasmic reticulum and around lipid droplets. Gain- and loss-of-function experiments have demonstrated that HILPDA promotes lipid storage in hepatocytes, macrophages and cancer cells. HILPDA increases lipid droplet accumulation at least partly by inhibiting triglyceride hydrolysis via ATGL and stimulating triglyceride synthesis via DGAT1. Overall, HILPDA is a novel regulatory signal that adjusts triglyceride storage and the intracellular availability of fatty acids to the external fatty acid supply and the capacity for oxidation.
Collapse
Affiliation(s)
- Montserrat A de la Rosa Rodriguez
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, the Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Stippeneng 4, 6708 WE, Wageningen, the Netherlands.
| |
Collapse
|
31
|
Matsumura K, Ito S. Novel biomarker genes which distinguish between smokers and chronic obstructive pulmonary disease patients with machine learning approach. BMC Pulm Med 2020; 20:29. [PMID: 32013930 PMCID: PMC6998147 DOI: 10.1186/s12890-020-1062-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
Background Chronic obstructive pulmonary disease (COPD) is combination of progressive lung diseases. The diagnosis of COPD is generally based on the pulmonary function testing, however, difficulties underlie in prognosis of smokers or early stage of COPD patients due to the complexity and heterogeneity of the pathogenesis. Computational analyses of omics technologies are expected as one of the solutions to resolve such complexities. Methods We obtained transcriptomic data by in vitro testing with exposures of human bronchial epithelial cells to the inducers for early events of COPD to identify the potential descriptive marker genes. With the identified genes, the machine learning technique was employed with the publicly available transcriptome data obtained from the lung specimens of COPD and non-COPD patients to develop the model that can reflect the risk continuum across smoking and COPD. Results The expression levels of 15 genes were commonly altered among in vitro tissues exposed to known inducible factors for earlier events of COPD (exposure to cigarette smoke, DNA damage, oxidative stress, and inflammation), and 10 of these genes and their corresponding proteins have not previously reported as COPD biomarkers. Although these genes were able to predict each group with 65% accuracy, the accuracy with which they were able to discriminate COPD subjects from smokers was only 29%. Furthermore, logistic regression enabled the conversion of gene expression levels to a numerical index, which we named the “potential risk factor (PRF)” index. The highest significant index value was recorded in COPD subjects (0.56 at the median), followed by smokers (0.30) and non-smokers (0.02). In vitro tissues exposed to cigarette smoke displayed dose-dependent increases of PRF, suggesting its utility for prospective risk estimation of tobacco products. Conclusions Our experimental-based transcriptomic analysis identified novel genes associated with COPD, and the 15 genes could distinguish smokers and COPD subjects from non-smokers via machine-learning classification with remarkable accuracy. We also suggested a PRF index that can quantitatively reflect the risk continuum across smoking and COPD pathogenesis, and we believe it will provide an improved understanding of smoking effects and new insights into COPD.
Collapse
Affiliation(s)
- Kazushi Matsumura
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan.
| | - Shigeaki Ito
- Scientific Product Assessment Center, R&D Group, Japan Tobacco Inc., 6-2 Umegaoka, Aoba-ku, Yokohama, Kanagawa, 227-8512, Japan
| |
Collapse
|
32
|
Kulminskaya N, Oberer M. Protein-protein interactions regulate the activity of Adipose Triglyceride Lipase in intracellular lipolysis. Biochimie 2020; 169:62-68. [DOI: 10.1016/j.biochi.2019.08.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/06/2019] [Indexed: 12/31/2022]
|
33
|
WY-14643 Regulates CYP1B1 Expression through Peroxisome Proliferator-Activated Receptor α-Mediated Signaling in Human Breast Cancer Cells. Int J Mol Sci 2019; 20:ijms20235928. [PMID: 31775380 PMCID: PMC6928855 DOI: 10.3390/ijms20235928] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/14/2022] Open
Abstract
Human cytochrome P450 1B1 (CYP1B1)-mediated biotransformation of endobiotics and xenobiotics plays an important role in the progression of human breast cancer. In this study, we investigated the effects of WY-14643, a peroxisome proliferator-activated receptor α (PPARα) agonist, on CYP1B1 expression and the related mechanism in MCF7 breast cancer cells. We performed quantitative reverse transcription-polymerase chain reaction, transient transfection, and chromatin immunoprecipitation to evaluate the effects of PPARα on peroxisome proliferator response element (PPRE)-mediated transcription. WY-14643 increased the protein and mRNA levels of CYP1B1, as well as promoter activity, in MCF-7 cells. Moreover, WY-14643 plus GW6471, a PPARα antagonist, significantly inhibited the WY-14643-mediated increase in CYP1B1 expression. PPARα knockdown by a small interfering RNA markedly suppressed the induction of CYP1B1 expression by WY-14643, suggesting that WY-14643 induces CYP1B1 expression via a PPARα-dependent mechanism. Bioinformatics analysis identified putative PPREs (−833/−813) within the promoter region of the CYP1B1 gene. Inactivation of these putative PPREs by deletion mutagenesis suppressed the WY-14643-mediated induction of CYP1B1 promoter activation. Furthermore, WY-14643 induced PPARα to assume a form capable of binding specifically to the PPRE-binding site in the CYP1B1 promoter. Our findings suggest that WY-14643 induces the expression of CYP1B1 through activation of PPARα.
Collapse
|
34
|
VandeKopple MJ, Wu J, Auer EN, Giaccia AJ, Denko NC, Papandreou I. HILPDA Regulates Lipid Metabolism, Lipid Droplet Abundance, and Response to Microenvironmental Stress in Solid Tumors. Mol Cancer Res 2019; 17:2089-2101. [PMID: 31308147 DOI: 10.1158/1541-7786.mcr-18-1343] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/24/2019] [Accepted: 07/10/2019] [Indexed: 01/05/2023]
Abstract
Accumulation of lipid droplets has been observed in an increasing range of tumors. However, the molecular determinants of this phenotype and the impact of the tumor microenvironment on lipid droplet dynamics are not well defined. The hypoxia-inducible and lipid droplet associated protein HILPDA is known to regulate lipid storage and physiologic responses to feeding conditions in mice, and was recently shown to promote hypoxic lipid droplet formation through inhibition of the rate-limiting lipase adipose triglyceride lipase (ATGL). Here, we identify fatty acid loading and nutrient deprivation-induced autophagy as stimuli of HILPDA-dependent lipid droplet growth. Using mouse embryonic fibroblasts and human tumor cells, we found that genetic ablation of HILPDA compromised hypoxia-fatty acid- and starvation-induced lipid droplet formation and triglyceride storage. Nutrient deprivation upregulated HILPDA protein posttranscriptionally by a mechanism requiring autophagic flux and lipid droplet turnover, independent of HIF1 transactivation. Mechanistically, loss of HILPDA led to elevated lipolysis, which could be corrected by inhibition of ATGL. Lipidomic analysis revealed not only quantitative but also qualitative differences in the glycerolipid and phospholipid profile of HILPDA wild-type and knockout cells, indicating additional HILPDA functions affecting lipid metabolism. Deletion studies of HILPDA mutants identified the N-terminal hydrophobic domain as sufficient for targeting to lipid droplets and restoration of triglyceride storage. In vivo, HILPDA-ablated cells showed decreased intratumoral triglyceride levels and impaired xenograft tumor growth associated with elevated levels of apoptosis. IMPLICATIONS: Tumor microenvironmental stresses induce changes in lipid droplet dynamics via HILPDA. Regulation of triglyceride hydrolysis is crucial for cell homeostasis and tumor growth.
Collapse
Affiliation(s)
- Matthew J VandeKopple
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jinghai Wu
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Erich N Auer
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Amato J Giaccia
- Department of Radiation Oncology, Stanford University, Stanford, California
| | - Nicholas C Denko
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Ioanna Papandreou
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| |
Collapse
|
35
|
Key Genes of Lipid Metabolism and WNT-Signaling Are Downregulated in Subcutaneous Adipose Tissue with Moderate Weight Loss. Nutrients 2019; 11:nu11030639. [PMID: 30884788 PMCID: PMC6471921 DOI: 10.3390/nu11030639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 03/12/2019] [Indexed: 02/07/2023] Open
Abstract
Smaller cross-sectional studies and bariatric surgery trials suggest that weight loss may change the expression of genes in adipose tissue that have been implicated in the development of metabolic diseases, but well-powered intervention trials are lacking. In post hoc analyses of data from a 12-week dietary intervention trial initially designed to compare metabolic effects of intermittent vs. continuous calorie restriction, we analyzed the effects of overall weight loss on the subcutaneous adipose tissue (SAT) transcriptome. Changes in the transcriptome were measured by microarray using SAT samples of 138 overweight or obese individuals (age range: 35–65 years, BMI range: 25–40, non-smokers, non-diabetics). Participants were grouped post hoc according to the degree of their weight loss by quartiles (average weight loss in quartiles 1 to 4: 0%, −3.2%, −5.9%, and −10.7%). Candidate genes showing differential expression with weight loss according to microarray analyses were validated by reverse transcription quantitative polymerase chain reaction (RT-qPCR), and fold changes (FCs) were calculated to quantify differences in gene expression. A comparison of individuals in the highest vs. the lowest weight loss quartile revealed 681 genes to be differentially expressed (corrected p < 0.05), with 40 showing FCs of at least 0.4. Out of these, expression changes in secreted frizzled-related protein 2 (SFRP2, FC = 0.65, p = 0.006), stearoyl-CoA desaturase (SCD, FC = −1.00, p < 0.001), and hypoxia inducible lipid droplet-associated (HILPDA, FC = −0.45, p = 0.001) with weight loss were confirmed by RT-qPCR. Dietary weight loss induces significant changes in the expression of genes implicated in lipid metabolism (SCD and HILPDA) and WNT-signaling (SFRP2) in SAT.
Collapse
|
36
|
Glantschnig C, Koenen M, Gil‐Lozano M, Karbiener M, Pickrahn I, Williams‐Dautovich J, Patel R, Cummins CL, Giroud M, Hartleben G, Vogl E, Blüher M, Tuckermann J, Uhlenhaut H, Herzig S, Scheideler M. A miR‐29a‐driven negative feedback loop regulates peripheral glucocorticoid receptor signaling. FASEB J 2019; 33:5924-5941. [DOI: 10.1096/fj.201801385rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Christina Glantschnig
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Mascha Koenen
- Institute of Comparative Molecular EndocrinologyUlm University Ulm Germany
| | - Manuel Gil‐Lozano
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Michael Karbiener
- Division of Phoniatrics, Speech, and SwallowingDepartment of OtorhinolaryngologyUniversity HospitalMedical University of Graz Graz Austria
| | - Ines Pickrahn
- Department of Legal MedicineUniversity of Salzburg Salzburg Austria
| | | | - Rucha Patel
- Department of Pharmaceutical SciencesUniversity of Toronto Toronto Ontario Canada
| | - Carolyn L. Cummins
- Department of Pharmaceutical SciencesUniversity of Toronto Toronto Ontario Canada
| | - Maude Giroud
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Götz Hartleben
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Elena Vogl
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Matthias Blüher
- Clinic for Endocrinology and NephrologyMedical Research Center Leipzig Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular EndocrinologyUlm University Ulm Germany
| | - Henriette Uhlenhaut
- Research Group Molecular EndocrinologyHelmholtz Center Munich Neuherberg Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
- School of MedicineTechnical University Munich Munich Germany
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| |
Collapse
|
37
|
Mu W, Wang Z, Zöller M. Ping-Pong-Tumor and Host in Pancreatic Cancer Progression. Front Oncol 2019; 9:1359. [PMID: 31921628 PMCID: PMC6927459 DOI: 10.3389/fonc.2019.01359] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Metastasis is the main cause of high pancreatic cancer (PaCa) mortality and trials dampening PaCa mortality rates are not satisfying. Tumor progression is driven by the crosstalk between tumor cells, predominantly cancer-initiating cells (CIC), and surrounding cells and tissues as well as distant organs, where tumor-derived extracellular vesicles (TEX) are of major importance. A strong stroma reaction, recruitment of immunosuppressive leukocytes, perineural invasion, and early spread toward the peritoneal cavity, liver, and lung are shared with several epithelial cell-derived cancer, but are most prominent in PaCa. Here, we report on the state of knowledge on the PaCIC markers Tspan8, alpha6beta4, CD44v6, CXCR4, LRP5/6, LRG5, claudin7, EpCAM, and CD133, which all, but at different steps, are engaged in the metastatic cascade, frequently via PaCIC-TEX. This includes the contribution of PaCIC markers to TEX biogenesis, targeting, and uptake. We then discuss PaCa-selective features, where feedback loops between stromal elements and tumor cells, including distorted transcription, signal transduction, and metabolic shifts, establish vicious circles. For the latter particularly pancreatic stellate cells (PSC) are responsible, furnishing PaCa to cope with poor angiogenesis-promoted hypoxia by metabolic shifts and direct nutrient transfer via vesicles. Furthermore, nerves including Schwann cells deliver a large range of tumor cell attracting factors and Schwann cells additionally support PaCa cell survival by signaling receptor binding. PSC, tumor-associated macrophages, and components of the dysplastic stroma contribute to perineural invasion with signaling pathway activation including the cholinergic system. Last, PaCa aggressiveness is strongly assisted by the immune system. Although rich in immune cells, only immunosuppressive cells and factors are recovered in proximity to tumor cells and hamper effector immune cells entering the tumor stroma. Besides a paucity of immunostimulatory factors and receptors, immunosuppressive cytokines, myeloid-derived suppressor cells, regulatory T-cells, and M2 macrophages as well as PSC actively inhibit effector cell activation. This accounts for NK cells of the non-adaptive and cytotoxic T-cells of the adaptive immune system. We anticipate further deciphering the molecular background of these recently unraveled intermingled phenomena may turn most lethal PaCa into a curatively treatable disease.
Collapse
Affiliation(s)
- Wei Mu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Wei Mu
| | - Zhe Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| | - Margot Zöller
- Department of Oncology, The First Affiliated Hospital of Guangdong, Pharmaceutical University, Guangzhou, China
| |
Collapse
|
38
|
Lee J, Lee J, Cho YS. Peroxisome Proliferator-Activated Receptor α Agonist and Its Target Nanog Cooperate to Induce Pluripotency. J Clin Med 2018; 7:jcm7120488. [PMID: 30486372 PMCID: PMC6306698 DOI: 10.3390/jcm7120488] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/22/2018] [Accepted: 11/23/2018] [Indexed: 01/23/2023] Open
Abstract
The pharmaceutical compounds that modulate pluripotent stem cell (PSC) identity and function are increasingly adopted to generate qualified PSCs and their derivatives, which have promising potential in regenerative medicine, in pursuit of more accuracy and safety and less cost. Here, we demonstrate the peroxisome proliferator-activated receptor α (PPARα) agonist as a novel enhancer of pluripotency acquisition and induced pluripotent stem cell (iPSC) generation. We found that PPARα agonist, examined and selected Food and Drug Administration (FDA) -approved compound libraries, increase the expression of pluripotency-associated genes, such as Nanog, Nr5A2, Oct4, and Rex1, during the reprogramming process and facilitate iPSC generation by enhancing their reprogramming efficiency. A reprogramming-promoting effect of PPARα occurred via the upregulation of Nanog, which is essential for the induction and maintenance of pluripotency. Through bioinformatic analysis, we identified putative peroxisome proliferator responsive elements (PPREs) located within the promoter region of the Nanog gene. We also determined that PPARα can activate Nanog transcription by specific binding to putative PPREs. Taken together, our findings suggest that PPARα is an important regulator of PSC pluripotency and reprogramming, and PPARα agonists can be used to improve PSC technology and regenerative medicine.
Collapse
Affiliation(s)
- Jungwoon Lee
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
| | - Jinhyuk Lee
- Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Biotechnology, KRIBB School, University of Science and Technology (UST), Daejeon 34113, Korea.
| | - Yee Sook Cho
- Stem Cell Research Laboratory, Immunotherapy Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Korea.
- Department of Bioscience, KRIBB School, University of Science and Technology (UST), Daejeon 34113, Korea.
| |
Collapse
|
39
|
Hypoxic Signaling and Cholesterol Lipotoxicity in Fatty Liver Disease Progression. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2548154. [PMID: 29955245 PMCID: PMC6000860 DOI: 10.1155/2018/2548154] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 02/06/2023]
Abstract
Cholesterol is the only lipid whose absorption in the gastrointestinal tract is limited by gate-keeping transporters and efflux mechanisms, preventing its rapid absorption and accumulation in the liver and blood vessels. In this review, I explored the current data regarding cholesterol accumulation in liver cells and key mechanisms in cholesterol-induced fatty liver disease associated with the activation of deleterious hypoxic and nitric oxide signal transduction pathways. Although nonalcoholic fatty liver disease (NAFLD) affects both obese and nonobese individuals, the mechanism of NAFLD progression in lean individuals with healthy metabolism is puzzling. Lean NAFLD individuals exhibit normal metabolic responses, implying that liver damage is not associated with impaired metabolism per se and that direct lipotoxic effects are crucial for disease progression. Several redox and oxidant signaling pathways involving cholesterol are at play in fatty liver disease development. These include impairment of the mitochondrial and lysosomal function by cholesterol loading of the inner-cell membranes; formation of cholesterol crystals and hepatocyte degradation; and crown-like structures surrounding degrading hepatocytes, activating Kupffer cells, and evoking inflammation. The current review focuses on the induction of liver inflammation, fibrosis, and steatosis by free cholesterol via the hypoxia-inducible factor 1α (HIF-1α), a main oxygen-sensing transcription factor involved in all stages of NAFLD. Cholesterol loading in hepatocytes can result in chronic HIF-1α activity because of the decreased oxygen availability and excessive production of nitric oxide and mitochondrial reactive oxygen species.
Collapse
|
40
|
Mao XG, Wang C, Liu DY, Zhang X, Wang L, Yan M, Zhang W, Zhu J, Li ZC, Mi C, Tian JY, Hou GD, Miao SY, Song ZX, Li JC, Xue XY. Hypoxia upregulates HIG2 expression and contributes to bevacizumab resistance in glioblastoma. Oncotarget 2018; 7:47808-47820. [PMID: 27329597 PMCID: PMC5216980 DOI: 10.18632/oncotarget.10029] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/17/2016] [Indexed: 01/08/2023] Open
Abstract
Hypoxia contributes to the maintenance of stem-like cells in glioblastoma (GBM), and activates vascular mimicry and tumor resistance to anti-angiogenesis treatments. The present study examined the expression patterns and biological significance of hypoxia-inducible protein 2 (HIG2, also known as HILPDA) in GBM. HIG2 was highly expressed in gliomas and was correlated with tumor grade, and high HIG2 expression independently predicted poor GBM patient prognosis. HIG2 was upregulated during hypoxia and by hypoxia mimics, and HIG2 knockdown in GBM cells inhibited cell proliferation and invasion. HIF1α bound to the HIG2 promoter and increased its expression in GBM cells, and HIG2 upregulated HIF1α expression. Reconstruction of a HIG2-related molecular network using bioinformatics methods revealed that HIG2 is closely correlated with angiogenesis genes, such as VEGFA, in GBM. HIG2 levels positively correlated with VEGFA in GBM samples. In addition, treatment of transplanted xenograft nude mice with bevacizumab (anti-angiogenesis therapy) resulted in HIG2 upregulation at late stages. We conclude that HIG2 is overexpressed in GBM and upregulated by hypoxia, and is a potential novel therapeutic target. HIG2 overexpression is an independent prognostic indicator and may promote tumor resistance to anti-angiogenesis treatments.
Collapse
Affiliation(s)
- Xing-Gang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Chao Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Dong-Ye Liu
- Northern Hospital, General Hospital of PLA Shenyang Military Area Command, Shenyang, Liaoning Province, People's Republic of China
| | - Xiang Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Ming Yan
- Department of Orthopaedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Wei Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Jun Zhu
- Department of Orthopedics, Changzheng Hospital, Second Military Medical University, Shanghai, People's Republic of China
| | - Zi-Chao Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Chen Mi
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Jing-Yang Tian
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Guang-Dong Hou
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Si-Yu Miao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Zi-Xuan Song
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Jin-Cheng Li
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| | - Xiao-Yan Xue
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
41
|
Padmanabha Das KM, Wechselberger L, Liziczai M, De la Rosa Rodriguez M, Grabner GF, Heier C, Viertlmayr R, Radler C, Lichtenegger J, Zimmermann R, Borst JW, Zechner R, Kersten S, Oberer M. Hypoxia-inducible lipid droplet-associated protein inhibits adipose triglyceride lipase. J Lipid Res 2018; 59:531-541. [PMID: 29326160 PMCID: PMC5832925 DOI: 10.1194/jlr.m082388] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 01/10/2018] [Indexed: 11/29/2022] Open
Abstract
Elaborate control mechanisms of intracellular triacylglycerol (TAG) breakdown are critically involved in the maintenance of energy homeostasis. Hypoxia-inducible lipid droplet-associated protein (HILPDA)/hypoxia-inducible gene-2 (Hig-2) has been shown to affect intracellular TAG levels, yet, the underlying molecular mechanisms are unclear. Here, we show that HILPDA inhibits adipose triglyceride lipase (ATGL), the enzyme catalyzing the first step of intracellular TAG hydrolysis. HILPDA shares structural similarity with G0/G1 switch gene 2 (G0S2), an established inhibitor of ATGL. HILPDA inhibits ATGL activity in a dose-dependent manner with an IC50 value of ∼2 μM. ATGL inhibition depends on the direct physical interaction of both proteins and involves the N-terminal hydrophobic region of HILPDA and the N-terminal patatin domain-containing segment of ATGL. Finally, confocal microscopy combined with Förster resonance energy transfer-fluorescence lifetime imaging microscopy analysis indicated that HILPDA and ATGL colocalize and physically interact intracellularly. These findings provide a rational biochemical explanation for the tissue-specific increased TAG accumulation in HILPDA-overexpressing transgenic mouse models.
Collapse
Affiliation(s)
| | - Lisa Wechselberger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Márton Liziczai
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | | | - Gernot F Grabner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Christoph Heier
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Roland Viertlmayr
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Claudia Radler
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Jörg Lichtenegger
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria.,BioTechMed-Graz, 8010 Graz, Austria
| | - Jan Willem Borst
- Laboratory of Biochemistry and Microspectroscopy Research Facility, Wageningen University, Wageningen, The Netherlands
| | - Rudolf Zechner
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria.,BioTechMed-Graz, 8010 Graz, Austria
| | - Sander Kersten
- Division of Human Nutrition University of Graz, 8010 Graz, Austria
| | - Monika Oberer
- Institute of Molecular Biosciences, University of Graz, 8010 Graz, Austria .,BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
42
|
de la Rosa Rodriguez MA, Kersten S. Regulation of lipid droplet-associated proteins by peroxisome proliferator-activated receptors. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1212-1220. [DOI: 10.1016/j.bbalip.2017.07.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/13/2017] [Accepted: 07/14/2017] [Indexed: 12/24/2022]
|
43
|
VandeKopple MJ, Wu J, Baer LA, Bal NC, Maurya SK, Kalyanasundaram A, Periasamy M, Stanford KI, Giaccia AJ, Denko NC, Papandreou I. Stress-responsive HILPDA is necessary for thermoregulation during fasting. J Endocrinol 2017; 235:27-38. [PMID: 28739822 PMCID: PMC5567683 DOI: 10.1530/joe-17-0289] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/24/2017] [Indexed: 02/02/2023]
Abstract
Hypoxia-inducible lipid droplet-associated protein (HILPDA) has been shown to localize to lipid droplets in nutrient-responsive cell types such as hepatocytes and adipocytes. However, its role in the control of whole-body homeostasis is not known. We sought to measure cell-intrinsic and systemic stress responses in a mouse strain harboring whole-body Hilpda deficiency. We generated a genetically engineered mouse model of whole-body HILPDA deficiency by replacing the coding Hilpda exon with luciferase. We subjected the knockout animals to environmental stresses and measured whole-animal metabolic and behavioral parameters. Brown adipocyte precursors were isolated and differentiated in vitro to quantify the impact of HILPDA ablation in lipid storage and mobilization in these cells. HILPDA-knockout animals are viable and fertile, but show reduced ambulatory activity and oxygen consumption at regular housing conditions. Acclimatization at thermoneutral conditions abolished the phenotypic differences observed at 22°C. When fasted, HILPDA KO mice are unable to maintain body temperature and become hypothermic at 22°C, without apparent abnormalities in blood chemistry parameters or tissue triglyceride content. HILPDA expression was upregulated during adipocyte differentiation and activation in vitro; however, it was not required for lipid droplet formation in brown adipocytes. We conclude that HILPDA is necessary for efficient fuel utilization suggesting a homeostatic role for Hilpda in sub-optimal environments.
Collapse
Affiliation(s)
| | - Jinghai Wu
- Department of Radiation OncologyThe Ohio State University, Columbus, Ohio, USA
| | - Lisa A Baer
- Dorothy M. Davis Heart and Lung Research InstituteDepartment of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA
| | - Naresh C Bal
- Department of Physiology and Cell BiologyThe Ohio State University, Columbus, Ohio, USA
| | - Santosh K Maurya
- Department of Physiology and Cell BiologyThe Ohio State University, Columbus, Ohio, USA
| | | | - Muthu Periasamy
- Department of Physiology and Cell BiologyThe Ohio State University, Columbus, Ohio, USA
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research InstituteDepartment of Physiology and Cell Biology, The Ohio State University, Columbus, Ohio, USA
| | - Amato J Giaccia
- Department of Radiation OncologyStanford University, Stanford, CA, USA
| | - Nicholas C Denko
- Department of Radiation OncologyThe Ohio State University, Columbus, Ohio, USA
| | - Ioanna Papandreou
- Department of Radiation OncologyThe Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
44
|
Maier A, Wu H, Cordasic N, Oefner P, Dietel B, Thiele C, Weidemann A, Eckardt KU, Warnecke C. Hypoxia-inducible protein 2 Hig2/Hilpda mediates neutral lipid accumulation in macrophages and contributes to atherosclerosis in apolipoprotein E-deficient mice. FASEB J 2017; 31:4971-4984. [PMID: 28760743 DOI: 10.1096/fj.201700235r] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 07/10/2017] [Indexed: 01/09/2023]
Abstract
Recently we identified hypoxia-inducible protein 2 (HIG2)/hypoxia-inducible lipid droplet-associated (HILPDA) as lipid droplet (LD) protein. Because HILPDA is highly expressed in atherosclerotic plaques, we examined its regulation and function in murine macrophages, compared it to the LD adipose differentiation-related protein (Adrp)/perilipin 2 (Plin2), and investigated its effects on atherogenesis in apolipoprotein E-deficient (ApoE-/-) mice. Tie2-Cre-driven Hilpda conditional knockout (cKO) did not affect viability, proliferation, and ATP levels in macrophages. Hilpda proved to be a target of hypoxia-inducible factor 1 (Hif-1) and peroxisome proliferator-activated receptors. In contrast, Adrp/Plin2 was not induced by Hif-1. Hilpda localized to the endoplasmic reticulum-LD interface, the site of LD formation. Hypoxic lipid accumulation and storage of oxidized LDL, cholesteryl esters and triglycerides were abolished in Hilpda cKO macrophages, independent of the glycolytic switch, fatty acid or lipoprotein uptake. Hilpda depletion reduced resistance against lipid overload and increased production of reactive oxygen species after reoxygenation. LPS-stimulated prostaglandin-E2 production was dysregulated in macrophages, demonstrating the substrate buffer and reservoir function of LDs for eicosanoid production. In ApoE-/- Hilpda cKO mice, total aortic plaque area, plaque macrophages and vascular Vegf expression were reduced. Thus, macrophage Hilpda is crucial to foam-cell formation and lipid deposition, and to controlled prostaglandin-E2 production. By these means Hilpda promotes lesion formation and progression of atherosclerosis.-Maier, A., Wu, H., Cordasic, N., Oefner, P., Dietel, B., Thiele, C., Weidemann, A., Eckardt, K.-U., Warnecke, C. Hypoxia-inducible protein 2 Hig2/Hilpda mediates neutral lipid accumulation in macrophages and contributes to atherosclerosis in apolipoprotein E-deficient mice.
Collapse
Affiliation(s)
- Anja Maier
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Hao Wu
- Department of Molecular Biology and Genetics, and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nada Cordasic
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Oefner
- Institute for Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Barbara Dietel
- Department of Molecular Cardiology and Angiology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Thiele
- Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; and
| | - Alexander Weidemann
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany.,Department of Medicine I, Nephrology, Transplantation, and Medical Intensive Care, University Witten/Herdecke, Medical Center Cologne-Merheim, Cologne, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christina Warnecke
- Department of Nephrology and Hypertension, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany;
| |
Collapse
|
45
|
ANGPTL4 promotes bile acid absorption during taurocholic acid supplementation via a mechanism dependent on the gut microbiota. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1056-1067. [PMID: 28733267 DOI: 10.1016/j.bbalip.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 07/06/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022]
Abstract
Angiopoietin-like 4 (ANGPTL4) raises plasma triglyceride levels by inhibiting lipoprotein lipase. A set of compounds that are able to reduce plasma triglyceride levels are bile acids (BA). Because BA have been shown to decrease ANGPTL4 secretion by intestinal cells, we hypothesized that BA lower plasma triglycerides (partly) via ANGPTL4. To test that hypothesis, wild-type and Angptl4-/- mice were fed chow supplemented with taurocholic acid (TCA) for seven days. TCA supplementation effectively lowered plasma triglycerides in wild-type and Angptl4-/- mice, indicating that ANGPTL4 is not required for plasma triglyceride-lowering by BA. Intriguingly, however, plasma and hepatic BA concentrations were significantly lower in TCA-supplemented Angptl4-/- mice than in TCA-supplemented wild-type mice. These changes in the Angptl4-/- mice were accompanied by lower BA levels in ileal scrapings and decreased expression of FXR-target genes in the ileum, including the BA transporter Slc10a2. By contrast, faecal excretion of specifically primary BA was higher in the Angptl4-/- mice, suggesting that loss of ANGPTL4 impairs intestinal BA absorption. Since the gut microbiota converts primary BA into secondary BA, elevated excretion of primary BA in Angptl4-/- mice may reflect differences in gut microbial composition and/or functionality. Indeed, colonic microbial composition was markedly different between Angptl4-/- and wild-type mice. Suppression of the gut bacteria using antibiotics abolished differences in plasma, hepatic, and faecal BA levels between TCA-supplemented Angptl4-/- and wild-type mice. In conclusion, 1) ANGPTL4 is not involved in the triglyceride-lowering effect of BA; 2) ANGPTL4 promotes BA absorption during TCA supplementation via a mechanism dependent on the gut microbiota.
Collapse
|
46
|
Meng XH, Chen B, Zhang JP. Intracellular Insulin and Impaired Autophagy in a Zebrafish model and a Cell Model of Type 2 diabetes. Int J Biol Sci 2017; 13:985-995. [PMID: 28924380 PMCID: PMC5599904 DOI: 10.7150/ijbs.19249] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 05/13/2017] [Indexed: 12/16/2022] Open
Abstract
Type 2 diabetes mellitus is characterized by insulin resistance. However, the complete molecular mechanism remains unclear. In this study, zebrafish were fed a long-term high-fat diet to induce type 2 diabetes, which resulted in a higher body weight, body mass index, more lipid vacuoles in liver, increased insulin transcription level in liver, brain and muscle, and high fasting blood glucose in the high-fat diet zebrafish. Oppositely, the transcription levels of insulin substrate-2 and glucose transporter 2 were significantly decreased, indicating insulin signaling pathway and glucose transport impaired in the insulin-targeting tissues. Transcription of the autophagy-related genes, ATG3, ATG4B, ATG5, ATG7, ATG12, and FOXO3, were decreased but autophagy inhibitor gene m-TOR increased, and autophagy-flux was inhibited in liver of the high-fat diet zebrafish. Main of these changes were confirmed in palmitic acid-treated HepG2 cells. Further, in co-immunoprecipitation and subcellular co-localization experiments, the conjunction of preproinsulin with cargo-recognition protein p62 increased, but conjuncts of autophagosome with p62-cargo, lysosomes with p62-cargo, and autolysosomes decreased apparently. Interestingly, lysosomes, autolysosomes and conjuncts of p62-insulin localized at the periphery of palmitic acid-treated cells, the margination of lysosomes may mediate deactivation of proteases activity. These findings suggest that intracellular high-lipid may trigger defective autophagy, defective downstream signaling of insulin and accumulated intracellular preproinsulin, leading to dysregulation of cell homeostasis mechanism, which may be one of reasons involved in insulin-resistance in type 2 diabetes.
Collapse
Affiliation(s)
- Xiang-Hui Meng
- Laboratory of pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Chen
- Laboratory of pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Pu Zhang
- Laboratory of pharmacology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Dijk W, Mattijssen F, de la Rosa Rodriguez M, Loza Valdes A, Loft A, Mandrup S, Kalkhoven E, Qi L, Borst JW, Kersten S. Hypoxia-Inducible Lipid Droplet-Associated Is Not a Direct Physiological Regulator of Lipolysis in Adipose Tissue. Endocrinology 2017; 158:1231-1251. [PMID: 28323980 PMCID: PMC5460841 DOI: 10.1210/en.2016-1809] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/13/2017] [Indexed: 12/20/2022]
Abstract
Triglycerides are stored in specialized organelles called lipid droplets. Numerous proteins have been shown to be physically associated with lipid droplets and govern their function. Previously, the protein hypoxia-inducible lipid droplet-associated (HILPDA) was localized to lipid droplets and was suggested to inhibit triglyceride lipolysis in hepatocytes. We confirm the partial localization of HILPDA to lipid droplets and show that HILPDA is highly abundant in adipose tissue, where its expression is controlled by the peroxisome proliferator-activated receptor γ and by β-adrenergic stimulation. Levels of HILPDA markedly increased during 3T3-L1 adipocyte differentiation. Nevertheless, silencing of Hilpda using small interfering RNA or overexpression of Hilpda using adenovirus did not show a clear impact on 3T3-L1 adipogenesis. Following β-adrenergic stimulation, the silencing of Hilpda in adipocytes did not significantly alter the release of nonesterified fatty acids (NEFA) and glycerol. By contrast, adenoviral-mediated overexpression of Hilpda modestly attenuated the release of NEFA from adipocytes following β-adrenergic stimulation. In mice, adipocyte-specific inactivation of Hilpda had no effect on plasma levels of NEFA and glycerol after fasting, cold exposure, or pharmacological β-adrenergic stimulation. In addition, other relevant metabolic parameters were unchanged by adipocyte-specific inactivation of Hilpda. Taken together, we find that HILPDA is highly abundant in adipose tissue, where its levels are induced by peroxisome proliferator-activated receptor γ and β-adrenergic stimulation. In contrast to the reported inhibition of lipolysis by HILPDA in hepatocytes, our data do not support an important direct role of HILPDA in the regulation of lipolysis in adipocytes in vivo and in vitro.
Collapse
Affiliation(s)
- Wieneke Dijk
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Frits Mattijssen
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Montserrat de la Rosa Rodriguez
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Angel Loza Valdes
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Anne Loft
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Susanne Mandrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Eric Kalkhoven
- Molecular Cancer Research and Center for Molecular Medicine, University Medical Centre Utrecht, 3584 CG Utrecht, The Netherlands
| | - Ling Qi
- University of Michigan Medical School, Ann Arbor, Michigan 48105
| | - Jan Willem Borst
- Laboratory of Biochemistry, Microspectroscopy Centre, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism, and Genomics Group, Division of Human Nutrition, Wageningen University, 6708 WE Wageningen, The Netherlands
- University of Michigan Medical School, Ann Arbor, Michigan 48105
| |
Collapse
|
48
|
Shin JH, Jung JH. Non-alcoholic fatty liver disease and flavonoids: Current perspectives. Clin Res Hepatol Gastroenterol 2017; 41:17-24. [PMID: 27545758 DOI: 10.1016/j.clinre.2016.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/21/2016] [Accepted: 07/07/2016] [Indexed: 02/04/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an accumulation of fat in the liver despite a low level of alcohol intake, with signs of hepatomegaly. Although in the past, NAFLD was predominantly viewed as an aspect of metabolic syndrome, it is now considered that it should be classified as an independent condition similar to obesity, diabetes, and hypertension. Therefore, new treatment strategies, not based on correcting insulin resistance, are needed for NAFLD. This work analyzes methods of prevention, therapeutic approaches, and mechanisms involved in NAFLD, focusing on the use of flavonoids (epigallocatechin-3-gallate, resveratrol, anthocyanins, and isoflavones) with high antioxidant capacity. In addition, the mechanisms of cholesterol accumulation in the liver are identified as potential avenues for entirely new approaches to NAFLD treatment, contrasting the well-known relation between neutral fat and NAFLD.
Collapse
Affiliation(s)
- Jung Hee Shin
- Department of Food & Nutrition, Joongbu University, 201 Daehak-ro, Chubu-myeon, Geumsan-gun, Chungcheongnam-Do, Republic of Korea.
| | - Ji Hye Jung
- Institute for Clinical Nutrition, Inje University, Mareunnae-ro 9, Jung-gu, Seoul, Republic of Korea.
| |
Collapse
|
49
|
Genomics of human fatty liver disease reveal mechanistically linked lipid droplet-associated gene regulations in bland steatosis and nonalcoholic steatohepatitis. Transl Res 2016; 177:41-69. [PMID: 27376874 DOI: 10.1016/j.trsl.2016.06.003] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 05/13/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common disorder hallmarked by excessive lipid deposits. Based on our recent research on lipid droplet (LD) formation in hepatocytes, we investigated LD-associated gene regulations in NAFLD of different grades, that is, steatosis vs steatohepatitis by comparing liver biopsies from healthy controls (N = 13) and NAFLD patients (N = 102). On average, more than 700 differentially expressed genes (DEGs) were identified of which 146 are mechanistically linked to LD formation. We identified 51 LD-associated DEGs frequently regulated in patient samples (range ≥5 to ≤102) with the liver-receptor homolog-1(NR5A2), that is, a key regulator of cholesterol metabolism being commonly repressed among 100 patients examined. With bland steatosis, notable regulations involved hypoxia-inducible lipid droplet-associated-protein and diacylglycerol-O-acyltransferase-2 renowned for their role in LD-growth. Conversely, nonalcoholic steatohepatitis-associated DEGs coded for epidermal growth factor receptor and TLR4 signaling with decreased expression of the GTPase Rab5 and the lipid phosphohydrolase PPAP2B thus highlighting adaptive responses to inflammation, LDL-mediated endocytosis and lipogenesis, respectively. Studies with steatotic primary human hepatocyte cultures demonstrated induction of LD-associated PLIN2, CIDEC, DNAAF1, whereas repressed expression of CPT1A, ANGPTL4, and PKLR informed on burdened mitochondrial metabolism. Equally, repressed expression of the B-lymphocyte chemoattractant CXCL13 and STAT4 as well as induced FGF21 evidenced amelioration of steatosis-related inflammation. In-vitro/in-vivo patient sample comparisons confirmed C-reactive protein, SOCS3, NR5A2, and SOD2 as commonly regulated. Lastly, STRING network analysis highlighted potential "druggable" targets with PLIN2, CIDEC, and hypoxia-inducible lipid droplet-associated-protein being confirmed by immunofluorescence microscopy. In conclusion, steatosis and steatohepatitis specific gene regulations informed on the pathogenesis of NAFLD to broaden the perspective of targeted therapies.
Collapse
|
50
|
Koizume S, Miyagi Y. Lipid Droplets: A Key Cellular Organelle Associated with Cancer Cell Survival under Normoxia and Hypoxia. Int J Mol Sci 2016; 17:ijms17091430. [PMID: 27589734 PMCID: PMC5037709 DOI: 10.3390/ijms17091430] [Citation(s) in RCA: 156] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 12/20/2022] Open
Abstract
The Warburg effect describes the phenomenon by which cancer cells obtain energy from glycolysis even under normoxic (O₂-sufficient) conditions. Tumor tissues are generally exposed to hypoxia owing to inefficient and aberrant vasculature. Cancer cells have multiple molecular mechanisms to adapt to such stress conditions by reprogramming the cellular metabolism. Hypoxia-inducible factors are major transcription factors induced in cancer cells in response to hypoxia that contribute to the metabolic changes. In addition, cancer cells within hypoxic tumor areas have reduced access to serum components such as nutrients and lipids. However, the effect of such serum factor deprivation on cancer cell biology in the context of tumor hypoxia is not fully understood. Cancer cells are lipid-rich under normoxia and hypoxia, leading to the increased generation of a cellular organelle, the lipid droplet (LD). In recent years, the LD-mediated stress response mechanisms of cancer cells have been revealed. This review focuses on the production and functions of LDs in various types of cancer cells in relation to the associated cellular environment factors including tissue oxygenation status and metabolic mechanisms. This information will contribute to the current understanding of how cancer cells adapt to diverse tumor environments to promote their survival.
Collapse
Affiliation(s)
- Shiro Koizume
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama 241-8515, Japan.
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama 241-8515, Japan.
| |
Collapse
|