1
|
Xiao S, Liu W, Zhang S, Schroyen M. The role of maternal dietary protein on livestock development, production and health. Anim Reprod Sci 2025; 276:107835. [PMID: 40199102 DOI: 10.1016/j.anireprosci.2025.107835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Maternal dietary protein plays a pivotal role in shaping offspring development, health, and productivity in economically important livestock species, including pigs, cattle, and sheep. Protein intake during gestation influences multiple physiological processes in the offspring, such as fetal growth, metabolic programming, muscle development, immune function, reproduction, and gut health. The specific effects of maternal protein intake vary depending on the species and the gestational period, as the demands for protein fluctuate throughout pregnancy to support fetal development and postnatal adaptation. This review systematically explores the effects of maternal protein nutrition on the offspring of different species and identifies the commonalities and differences observed in the studies. Studies indicate that maternal protein restriction can lead to lower birth weights, impaired muscle growth, altered metabolic programming, and compromised immune function in offspring, potentially affecting their long-term productivity. Conversely, excessive protein intake may also have adverse effects, such as immune dysregulation and metabolic imbalances. The impact of maternal protein levels extends beyond birth, influencing postnatal growth trajectories, reproductive performance, and gut microbiota composition. While considerable progress has been made in understanding these relationships, gaps remain in identifying the precise molecular mechanisms underlying these effects. Future research should focus on refining dietary recommendations tailored to different livestock species, investigating the role of gestation stage-specific protein requirements, and integrating multi-omics approaches to elucidate the long-term consequences of maternal protein intake. A deeper understanding of these mechanisms will contribute to optimizing feeding strategies, enhancing animal welfare, and improving the sustainability of livestock production systems.
Collapse
Affiliation(s)
- Shize Xiao
- Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés 2, Gembloux 5030, Belgium
| | - Wenju Liu
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, HuaZhong Agricultural University, Wuhan, China
| | - Shujun Zhang
- Key Lab of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education, HuaZhong Agricultural University, Wuhan, China
| | - Martine Schroyen
- Gembloux Agro-Bio Tech, University of Liège, Passage des Déportés 2, Gembloux 5030, Belgium.
| |
Collapse
|
2
|
Dargham T, Aguilera-Correa JJ, Avellan R, Mallick I, Celik L, Santucci P, Brasseur G, Poncin I, Point V, Audebert S, Camoin L, Daher W, Cavalier JF, Kremer L, Canaan S. A proteomic and functional view of intrabacterial lipid inclusion biogenesis in mycobacteria. mBio 2025; 16:e0147524. [PMID: 39998225 PMCID: PMC11980559 DOI: 10.1128/mbio.01475-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 02/03/2025] [Indexed: 02/26/2025] Open
Abstract
During infection and granuloma formation, pathogenic mycobacteria store triacylglycerol as intrabacterial lipid inclusions (ILIs). This accumulation of nutrients provides a carbon source for bacterial persistence and slows down intracellular metabolism. Mycobacterium abscessus (Mab), a rapidly growing non-tuberculous actinobacterium, produces ILI throughout its infection cycle. Here, Mab was used as a model organism to identify proteins associated with ILI accumulation on a global scale. By using the APEX2 proximity labeling method in an in vitro model for ILI accumulation, we identified 228 proteins possibly implicated in ILI biosynthesis. Fluorescence microscopy of strains overexpressing eight ILI-associated proteins (IAP) candidates fused to superfolder green fluorescent protein showed co-localization with ILI. Genetic inactivation of these potential IAP-encoding genes and subsequent lipid analysis emphasized the importance of MAB_3486 and MAB_4532c as key enzymes influencing triacylglycerol storage. This study underscores the dynamic process of ILI biogenesis and advances our understanding of lipid metabolism in pathogenic mycobacteria. Identifying major IAP in lipid accumulation offers new therapeutic perspectives to control the growth and persistence of pathogenic mycobacteria. IMPORTANCE This study sheds light into the complex process of intracellular lipid accumulation and storage in the survival and persistence of pathogenic mycobacteria, which is of clinical relevance. By identifying the proteins involved in the formation of intrabacterial lipid inclusions and revealing their impact on lipid metabolism, our data may lead to the development of new therapeutic strategies to target and control pathogenic mycobacteria, potentially improving outcomes for patients with mycobacterial infections.
Collapse
Affiliation(s)
- Tonia Dargham
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
- IHU Méditerranée Infection, Aix-Marseille Univ., Marseille, France
| | | | - Romain Avellan
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Ivy Mallick
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Léa Celik
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Pierre Santucci
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Gael Brasseur
- Aix-Marseille Univ., CNRS, LCB-UMR 7283, IMM FR3479, IM2B, Marseille, France
| | - Isabelle Poncin
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Vanessa Point
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| | - Stéphane Audebert
- Aix-Marseille Univ., INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Luc Camoin
- Aix-Marseille Univ., INSERM, CNRS, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Wassim Daher
- CNRS UMR 9004, IRIM, Université de Montpellier, Montpellier, France
- INSERM, IRIM, Montpellier, France
| | | | - Laurent Kremer
- CNRS UMR 9004, IRIM, Université de Montpellier, Montpellier, France
- INSERM, IRIM, Montpellier, France
| | - Stéphane Canaan
- Aix-Marseille Univ., CNRS, LISM-UMR 7255, IMM FR3479, IM2B, Marseille, France
| |
Collapse
|
3
|
Liu Z, Peng H, Liu P, Duan F, Yang Y, Li P, Li Z, Wu J, Chang J, Shang D, Tian Q, Zhang J, Xie Y, Liu Z, An Y. Deciphering significances of autophagy in the development and metabolism of adipose tissue. Exp Cell Res 2025; 446:114478. [PMID: 39978716 DOI: 10.1016/j.yexcr.2025.114478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/17/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
The mechanisms of adipose tissue activation and inactivation have been a hot topic of research in the last decade, from which countermeasures have been attempted to be found against obesity as well as other lipid metabolism-related diseases, such as type 2 diabetes mellitus and non-alcoholic fatty liver disease. Autophagy has been shown to be closely related to the regulation of adipocyte activity, which is involved in the whole process including white adipocyte differentiation/maturation and brown or beige adipocyte generation/activation. Dysregulation of autophagy in adipose tissue has been demonstrated to be associated with obesity. On this basis, we summarize the pathways and mechanisms of autophagy involved in the regulation of lipid metabolism and present a review of its pathophysiological roles in lipid metabolism-related diseases, in the hope of providing ideas for the treatment of these diseases.
Collapse
Affiliation(s)
- Zitao Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Haoyuan Peng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengfei Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Feiyi Duan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yutian Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Pengkun Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Zhihao Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiaoyan Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Jiayi Chang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Dandan Shang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Qiwen Tian
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Jiawei Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Yucheng Xie
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; School of Stomatology, Henan University, Kaifeng, 475004, China
| | - Zhenzhen Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, China; Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, 475004, China; Henan Provincial Research Center of Engineering Technology for Nuclear Protein Medical Detection, Zhengzhou Health College, Zhengzhou, 450064, China.
| |
Collapse
|
4
|
Valdés-Hernández J, Ramayo-Caldas Y, Passols M, Criado-Mesas L, Castelló A, Sánchez A, Folch JM. Identification of differentially expressed genes and polymorphisms related to intramuscular oleic-to-stearic fatty acid ratio in pigs. Anim Genet 2025; 56:e13491. [PMID: 39593270 DOI: 10.1111/age.13491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 10/11/2024] [Accepted: 11/06/2024] [Indexed: 11/28/2024]
Abstract
The intramuscular oleic-to-stearic fatty acid ratio (C18:1n-9/C18:0) is an important indicator of the biosynthesis and desaturation of fatty acids in muscle. By using an RNA-Seq approach in muscle samples from 32 BC1_DU (25% Iberian and 75% Duroc) pigs with divergent values (high: H and low: L) of C18:1n-9/C18:0 fatty acids ratio, a total of 81 differentially expressed genes (DEGs) were identified. Functional analyses of DEGs indicate that mainly peroxisome proliferator-activated receptor signaling pathway (associated genes: PPARG, SCD, PLIN1, and FABP3) was overrepresented. Notably, SCD is directly involved in the conversion of C18:0 to C18:1n-9, and PPARG is a transcription factor regulating lipid metabolism genes, including SCD. However, other DEGs (e.g., ACADVL, FADS3, EPHB2, HGFAC, NGFR, NR0B2, MDH1, MMAA, PPP1R1B, SFRP5, RAB30, and TRARG1) are plausible candidate genes to explain the phenotypic differences of the C18:1n-9/C18:0 ratio. Interestingly, seven genetic variants within the SCD (including the well-known AY487830:g.2228T>C SNP and other novel genotyped polymorphisms) are associated with two haplotypes. Although the haplotypes are segregating at different frequencies in the H and L groups, they do not fully explain the desaturation ratios or the SCD expression levels. A more complex model, including polyunsaturated fatty acids such as C18:2n-6, C20:4n-6, and C18:3n-3, is suggested to explain the regulation of the C18:1n-9/C18:0 desaturation ratio in porcine muscle.
Collapse
Affiliation(s)
- Jesús Valdés-Hernández
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i Dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Yuliaxis Ramayo-Caldas
- Departament de Genètica i Millora Animal, Institut de Recerca y Tecnologia Agraroalimentàries (IRTA), Caldes de Montbui, Spain
| | - Magí Passols
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i Dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Lourdes Criado-Mesas
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i Dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Anna Castelló
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i Dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Armand Sánchez
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i Dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| | - Josep M Folch
- Plant and Animal Genomics, Centre for Research in Agricultural Genomics (CRAG), CSIC-IRTA-UAB-UB, Campus UAB, Bellaterra, Spain
- Departament de Ciència Animal i Dels Aliments, Facultat de Veterinària, Universitat Autònoma de Barcelona (UAB), Bellaterra, Spain
| |
Collapse
|
5
|
Cui N, Lei T, Liang L, Zhou S, Jin X, Shi Y, Zhao Z, Song X. Type I interferon (IFNd) enhanced the innate immune response and lipid droplets (LDs) formation in obscure puffer Takifugu obscurus. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 163:105321. [PMID: 39855437 DOI: 10.1016/j.dci.2025.105321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/03/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
As one member of type I IFN, IFNd showed huge distinctive responses and activity during the viral or bacterial infection in various fish species. Our previous study identified IFNd from obscure puffer Takifugu obscurus (ToIFNd), and the function and regulation of ToIFNd was further investigated in the present study. The transcriptional levels of ToIFNd were significantly induced post bacteria Vibrio harveyi and virus simulator Poly (I:C) stimulation in the head kidney. In addition, recombinant ToIFNd (rToIFNd) treatment enhanced the expressions of numerous interferon stimulated genes (ISGs, such as Mx1, PKR1, and PKR2) and several immune effectors (such as Il1, faslg, and tnf14) both in the in vivo and in vitro experiments. The expression patterns of interferon regulatory factors (IRFs) post rToIFNd stimulation suggesting that IRF3, 7, and 8 showed strong response and might play critical role during the ToIFNd mediated signal transduction. Remarkably, we firstly found that rToIFNd stimulation promoted lipid droplets (LDs) formation in liver, which was further confirmed by the increased number of LDs by TEM, up-regulated expressions of LDs marker PLIN3, as well as the transcripts of genes involved in the transport and synthesis of neutral lipids. These results corroborate the antibacterial and antiviral function of ToIFNd in obscure puffer, and revealed that ToIFNd might play immune regulatory role by medicating LDs formation, which will contribute to explore the functional characteristics of IFN system.
Collapse
Affiliation(s)
- Nan Cui
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Tianying Lei
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Lanyue Liang
- Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Shan Zhou
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Xingkun Jin
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Yan Shi
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Zhe Zhao
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China
| | - Xiaorui Song
- Jiangsu Province Engineering Research Center for Marine Bio-resources Sustainable Utilization, Hohai University, Nanjing, 210024, China; Department of Marine Biology, College of Oceanography, Hohai University, Nanjing, 210024, China.
| |
Collapse
|
6
|
Liang B, Fu L, Liu P. Regulation of lipid droplet dynamics and lipid homeostasis by hydroxysteroid dehydrogenase proteins. Trends Cell Biol 2025; 35:153-165. [PMID: 39603915 DOI: 10.1016/j.tcb.2024.10.010] [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: 07/17/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
The superfamily of hydroxysteroid dehydrogenases (HSDs) has been well-characterized as enzymes in lipid metabolism, and especially in steroid hormone metabolism from bacteria to mammals. Recently, a subset of HSDs members, including 3β-HSD, 11β-HSD, and 17β-HSD, have been shown to be lipid droplet (LD)-associated proteins that are involved in LD dynamics beyond their canonical functions. This review summarizes current understanding of these LD-associated HSD proteins, focusing on how they regulate different LDs with respect to distinct neutral lipids including triacylglycerols (TAGs), cholesterol esters (CEs), and retinyl esters (REs), the evolutionally conserved role of some LD-associated 17β-HSDs in preventing lipolysis, and specific targeting of HSDs for the treatment of metabolic diseases and viral infections.
Collapse
Affiliation(s)
- Bin Liang
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming 650500, China; Southwest United Graduate School, Kunming 650092, China.
| | - Lin Fu
- Center for Life Sciences, Yunnan Key Laboratory of Cell Metabolism and Diseases, School of Life Sciences, Yunnan University, Kunming 650500, China; Key Laboratory of Tumor Immunological Prevention and Treatment in Yunnan Province, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan 650051, China.
| | - Pingsheng Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of the Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
7
|
He H, Lin Y, Zhang X, Xie H, Wang Z, Hu S, Li L, Liu H, Han C, Xia L, Hu J, Wang J, Liao L, Yuan X. Transcriptome Analysis Reveals the Molecular Mechanism of PLIN1 in Goose Hierarchical and Pre-Hierarchical Follicle Granulosa Cells. Animals (Basel) 2025; 15:284. [PMID: 39858284 PMCID: PMC11761271 DOI: 10.3390/ani15020284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/06/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
PLIN1, a member of the PAT family, is expressed in both adipocytes and steroidogenic cells. In this study, we used cell transfection technology combined with transcriptome sequencing to investigate the regulatory mechanism of PLIN1 in goose follicular GCs. Gene Ontology (GO) analysis revealed that in the four groups (phGC: over_vs_over-NC; hGC: over_vs_over-NC; phGC: si_vs_si-NC; hGC: si_vs_si-NC), most differentially expressed genes (DEGs) were significantly enriched (p < 0.05) in pathways related to biological processes (BPs), particularly those associated with the regulation of cellular lipid metabolism and oxidative stress. KEGG analysis further identified significant enrichment (p < 0.05) in pathways related to cell apoptosis and the cell cycle. A joint analysis of KEGG and PPI on the upregulated and downregulated DEGs revealed that the TGF-β signaling pathway was the only pathway significantly enriched among both upregulated and downregulated DEGs after PLIN1 overexpression in hGCs and phGCs. Based on these findings, we hypothesize that PLIN1 overexpression may promote granulosa cell proliferation and apoptosis by activating the TGF-β signaling pathway in goose follicular GCs. Additionally, nine potential candidate genes were identified: PPARγ, MGLL, PTEN, BAMBI, BMPR2, JUN, FST, ACSF3, and ACSL4. These results address a significant research gap concerning the role of this gene in granulosa cells and contribute to the understanding of its molecular regulatory mechanisms.
Collapse
Affiliation(s)
- Hua He
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Yueyue Lin
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Xi Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Hengli Xie
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Zhujun Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Hehe Liu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Chunchun Han
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Lu Xia
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Jiwei Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| | - Lin Liao
- College of Horticulture, Sichuan Agricultural University, Chengdu 611130, China
| | - Xin Yuan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China; (H.H.); (Y.L.)
| |
Collapse
|
8
|
de Miranda RPR, Soares TKDA, Castro DP, Genta FA. General aspects, host interaction, and application of Metarhizium sp. in arthropod pest and vector control. FRONTIERS IN FUNGAL BIOLOGY 2024; 5:1456964. [PMID: 39634290 PMCID: PMC11614621 DOI: 10.3389/ffunb.2024.1456964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 10/14/2024] [Indexed: 12/07/2024]
Abstract
The application of microorganisms as bio-control agents against arthropod populations is a need in many countries, especially in tropical, subtropical, and neotropical endemic areas. Several arthropod species became agricultural pests of paramount economic significance, and many methods have been developed for field and urban applications to prevent their, the most common being the application of chemical insecticides. However, the indiscriminate treatment based upon those substances acted as a selective pressure for upcoming resistant phenotype populations. As alternative tools, microorganisms have been prospected as complementary tools for pest and vectorial control, once they act in a more specific pattern against target organisms than chemicals. They are considered environmentally friendly since they have considerably less off-target effects. Entomopathogenic fungi are organisms capable of exerting pathogenesis in many vector species, thus becoming potential tools for biological management. The entomopathogenic fungi Metarhizium sp. have been investigated as a microbiological agent for the control of populations of insects in tropical regions. However, the development of entomopathogenic fungi as control tools depends on physiological studies regarding aspects such as mechanisms of pathogenicity, secreted enzymes, viability, and host-pathogen aspects. The following review briefly narrates current aspects of entomopathogenic fungi, such as physiology, cellular characteristics, host-pathogen interactions, and its previous applications against different insect orders with medical and economic importance. Approaches integrating new isolation, prospection, characterization, delivery strategies, formulations, and molecular and genetic tools will be decisive to elucidate the molecular mechanisms of EPFs and to develop more sustainable alternative pesticides.
Collapse
Affiliation(s)
| | | | - Daniele Pereira Castro
- Laboratorio de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| | - Fernando Ariel Genta
- Laboratorio de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Lai TT, Ishida M, Kosaka H, Matsui K, Matsushima H, Yamamoto H, Kiguchi G, Nguyen KV, Inoue K, Takada M, Kato H, Hirose Y, Yoshii K, Kaibori M. The Prognostic Impact of Adipophilin Expression on Long-Term Survival Following Liver Resection in Patients with Colorectal Liver Metastases. Cancers (Basel) 2024; 16:3827. [PMID: 39594782 PMCID: PMC11592894 DOI: 10.3390/cancers16223827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/10/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Background/Objectives: Adipophilin (ADP) is a protein associated with lipid droplets, and its expression is related to poor prognosis in certain cancers. However, its impact on the survival of patients with colorectal liver metastases (CRLMs) remains unclear. This study investigated the impact of ADP expression on long-term survival following hepatectomy in patients with CRLM. Methods: We retrospectively analyzed 102 consecutive patients who underwent hepatectomy between 2006 and 2022. ADP expression was examined in resected specimens through immunohistochemical staining using tissue microarrays. Long-term outcomes for ADP-positive (n = 51) and ADP-negative (n = 51) groups were compared with Kaplan-Meier survival analysis. Results: We found significantly decreased 5-year recurrence-free survival (RFS) and overall survival (OS) rates for ADP-positive patients relative to ADP-negative patients (29.4% versus 52.1%, respectively; p = 0.001 and 43.7% versus 72.2%, respectively; p = 0.003). Moreover, multivariate Cox hazards analysis demonstrated that patients with ADP-positive CRLM had a worse prognosis after hepatectomy than those with ADP-negative CRLM, as reflected by both RFS (HR 2.46, 95% CI 1.39-4.36, p = 0.002) and OS (HR: 2.89, 95% CI 1.43-5.85, p = 0.003). Conclusions: ADP expression had a significant prognostic impact on the survival of patients with CRLM following liver resection and may aid in optimal treatment planning.
Collapse
Affiliation(s)
- Tung Thanh Lai
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
- Department of Surgery, Hanoi Medical University, Hanoi 100000, Vietnam
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (M.I.); (Y.H.)
| | - Hisashi Kosaka
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| | - Kosuke Matsui
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| | - Hideyuki Matsushima
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| | - Hidekazu Yamamoto
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| | - Gozo Kiguchi
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| | - Khanh Van Nguyen
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
- Internal Gastroenterology Department, VNU University of Medicine and Pharmacy, Hanoi 100000, Vietnam
| | - Kyoko Inoue
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| | - Moriyasu Takada
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| | - Hiroki Kato
- Department of Mathematics and Statistics in Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (H.K.); (K.Y.)
| | - Yoshinobu Hirose
- Department of Pathology, Osaka Medical and Pharmaceutical University, Osaka 569-8686, Japan; (M.I.); (Y.H.)
| | - Kengo Yoshii
- Department of Mathematics and Statistics in Medical Sciences, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan; (H.K.); (K.Y.)
| | - Masaki Kaibori
- Department of Hepatobiliary Surgery, Kansai Medical University, Osaka 573-1010, Japan; (T.T.L.); (H.K.); (K.M.); (H.M.); (H.Y.); (G.K.); (K.V.N.); (K.I.); (M.T.)
| |
Collapse
|
10
|
Zhang C, Zheng M, Bai R, Chen J, Yang H, Luo G. Molecular mechanisms of lipid droplets-mitochondria coupling in obesity and metabolic syndrome: insights and pharmacological implications. Front Physiol 2024; 15:1491815. [PMID: 39588271 PMCID: PMC11586377 DOI: 10.3389/fphys.2024.1491815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/29/2024] [Indexed: 11/27/2024] Open
Abstract
Abnormal lipid accumulation is a fundamental contributor to obesity and metabolic disorders. Lipid droplets (LDs) and mitochondria (MT) serve as organelle chaperones in lipid metabolism and energy balance. LDs play a crucial role in lipid storage and mobilization, working in conjunction with MT to regulate lipid metabolism within the liver, brown adipose tissue, and skeletal muscle, thereby maintaining metabolic homeostasis. The novelty of our review is the comprehensive description of LD and MT interaction mechanisms. We also focus on the current drugs that target this metabolism, which provide novel approaches for obesity and related metabolism disorder treatment.
Collapse
Affiliation(s)
- Chunmei Zhang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mingxuan Zheng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Runlin Bai
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiale Chen
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Yang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Gan Luo
- Department of Orthopedics, Chengdu Integrated Traditional Chinese Medicine & Western Medicine Hospital/Chengdu First People’s Hospital, Chengdu, China
| |
Collapse
|
11
|
Kong L, Bai Q, Li C, Wang Q, Wang Y, Shao X, Wei Y, Sun J, Yu Z, Yin J, Shi B, Fang H, Chen X, Chen Q. Molecular probes for tracking lipid droplet membrane dynamics. Nat Commun 2024; 15:9413. [PMID: 39482302 PMCID: PMC11528070 DOI: 10.1038/s41467-024-53667-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 10/16/2024] [Indexed: 11/03/2024] Open
Abstract
Lipid droplets (LDs) feature a unique monolayer lipid membrane that has not been extensively studied due to the lack of suitable molecular probes that are able to distinguish this membrane from the LD lipid core. In this work, we present a three-pronged molecular probe design strategy that combines lipophilicity-based organelle targeting with microenvironment-dependent activation and design an LD membrane labeling pro-probe called LDM. Upon activation by the HClO/ClO- microenvironment that surrounds LDs, LDM pro-probe releases LDM-OH probe that binds to LD membrane proteins thus enabling visualization of the ring-like LD membrane. By utilizing LDM, we identify the dynamic mechanism of LD membrane contacts and their protein accumulation parameters. Taken together, LDM represents the first molecular probe for imaging LD membranes in live cells to the best of our knowledge, and represents an attractive tool for further investigations into the specific regulatory mechanisms with LD-related metabolism diseases and drug screening.
Collapse
Affiliation(s)
- Lingxiu Kong
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Qingjie Bai
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Cuicui Li
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore
| | - Qiqin Wang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
- College of Pharmacy, Jinan University, Guangzhou, 510632, PR China
| | - Yanfeng Wang
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Xintian Shao
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
- Neck-Shoulder and Lumbocrural Pain Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Yongchun Wei
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Jiarao Sun
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Zhenjie Yu
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Junling Yin
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Bin Shi
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
- Neck-Shoulder and Lumbocrural Pain Hospital, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China
| | - Hongbao Fang
- College of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, PR China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Qixin Chen
- School of Pharmaceutical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China.
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, PR China.
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, Singapore, 138667, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| |
Collapse
|
12
|
Paris JR, Nitta Fernandes FA, Pirri F, Greco S, Gerdol M, Pallavicini A, Benoiste M, Cornec C, Zane L, Haas B, Le Bohec C, Trucchi E. Gene Expression Shifts in Emperor Penguin Adaptation to the Extreme Antarctic Environment. Mol Ecol 2024:e17552. [PMID: 39415606 DOI: 10.1111/mec.17552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 09/17/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
Gene expression can accelerate ecological divergence by rapidly tweaking the response of an organism to novel environments, with more divergent environments exerting stronger selection and supposedly, requiring faster adaptive responses. Organisms adapted to extreme environments provide ideal systems to test this hypothesis, particularly when compared to related species with milder ecological niches. The Emperor penguin (Aptenodytes forsteri) is the only endothermic vertebrate breeding in the harsh Antarctic winter, in stark contrast with the less cold-adapted sister species, the King penguin (A. patagonicus). Assembling the first de novo transcriptomes and analysing multi-tissue (brain, kidney, liver, muscle, skin) RNA-Seq data from natural populations of both species, we quantified the shifts in tissue-enhanced genes, co-expression gene networks, and differentially expressed genes characterising Emperor penguin adaptation to the extreme Antarctic. Our analyses revealed the crucial role played by muscle and liver in temperature homeostasis, fasting, and whole-body energy metabolism (glucose/insulin regulation, lipid metabolism, fatty acid beta-oxidation, and blood coagulation). Repatterning at the regulatory level appears as more important in the brain of the Emperor penguin, showing the lowest signature of differential gene expression, but the largest co-expression gene network shift. Nevertheless, over-expressed genes related to mTOR signalling in the brain and the liver support their central role in cold and fasting responses. Besides contributing to understanding the genetics underlying complex traits, like body energy reservoir management, our results provide a first insight into the role of gene expression in adaptation to one of the most extreme environmental conditions endured by an endotherm.
Collapse
Affiliation(s)
- Josephine R Paris
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Flávia A Nitta Fernandes
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
| | - Federica Pirri
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
- Department of Biology, University of Padova, Padova, Italy
| | - Samuele Greco
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Marco Gerdol
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Marine Benoiste
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
| | - Clément Cornec
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
- ENES Bioacoustics Research Laboratory, CRNL, CNRS, Inserm, University of Lyon, Saint-Etienne, France
| | - Lorenzo Zane
- Department of Biology, University of Padova, Padova, Italy
| | - Brian Haas
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Céline Le Bohec
- Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg, France
- CEFE, Université de Montpellier, CNRS, EPHE, IRD, Montpellier, France
- Département de Biologie Polaire, Centre Scientifique de Monaco, Monaco, Monaco
| | - Emiliano Trucchi
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| |
Collapse
|
13
|
Wang Y, Zhou J, Yang Q, Li X, Qiu Y, Zhang Y, Liu M, Zhu AJ. Therapeutic siRNA targeting PLIN2 ameliorates steatosis, inflammation, and fibrosis in steatotic liver disease models. J Lipid Res 2024; 65:100635. [PMID: 39187042 PMCID: PMC11440260 DOI: 10.1016/j.jlr.2024.100635] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/10/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is the most prevalent chronic liver disease worldwide. If left untreated, MASLD can progress from simple hepatic steatosis to metabolic dysfunction-associated steatohepatitis, which is characterized by inflammation and fibrosis. Current treatment options for MASLD remain limited, leaving substantial unmet medical needs for innovative therapeutic approaches. Here, we show that PLIN2, a lipid droplet protein inhibiting hepatic lipolysis, serves as a promising therapeutic target for MASLD. Hepatic PLIN2 levels were markedly elevated in multiple MASLD mouse models induced by diverse nutritional and genetic factors. The liver-specific deletion of Plin2 exhibited significant anti-MASLD effects in these models. To translate this discovery into a therapeutic application, we developed a GalNAc-siRNA conjugate with enhanced stabilization chemistry and validated its potent and sustained efficacy in suppressing Plin2 expression in mouse livers. This siRNA therapeutic, named GalNAc-siPlin2, was shown to be biosafe in mice. Treatment with GalNAc-siPlin2 for 6-8 weeks led to a decrease in hepatic triglyceride levels by approximately 60% in high-fat diet- and obesity-induced MASLD mouse models, accompanied with increased hepatic secretion of VLDL-triglyceride and enhanced thermogenesis in brown adipose tissues. Eight-week treatment with GalNAc-siPlin2 significantly improved hepatic steatosis, inflammation, and fibrosis in high-fat/high fructose-induced metabolic dysfunction-associated steatohepatitis models compared to control group. As a proof of concept, we developed a GalNAc-siRNA therapeutic targeting human PLIN2, which effectively suppressed hepatic PLIN2 expression and ameliorated MASLD in humanized PLIN2 knockin mice. Together, our results highlight the potential of GalNAc-siPLIN2 as a candidate MASLD therapeutic for clinical trials.
Collapse
Affiliation(s)
- Yao Wang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Jiaxin Zhou
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Qi Yang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China
| | - Xinmeng Li
- Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yifu Qiu
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; Institute of Molecular Medicine, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, College of Future Technology, Peking University, Beijing, China
| | - Yansong Zhang
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, China.
| | - Min Liu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China.
| | - Alan Jian Zhu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Antoñanzas J, Núñez-Córdoba JM, Salido-Vallejo R, Álvarez-Gigli L, Robledano R, España A. Association between adipophilin expression and risk of dyslipidaemia in patients with granuloma annulare. Med Clin (Barc) 2024; 163:232-237. [PMID: 38853069 DOI: 10.1016/j.medcli.2024.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 06/11/2024]
Abstract
BACKGROUND An association between granuloma annulare (GA) and dyslipidaemia has been reported. Adipophilin expression may play a plausible role as a cutaneous biomarker for dyslipidaemia in patients with GA; however, this potential link remains to be explored. METHODS Patients with GA were identified at our hospital between January 1, 1990, and December 31, 2021, with a thorough review of their clinical and histological characteristics. Adipophilin staining was assessed in biopsies of GA lesions. RESULTS A total of 107 patients with GA were included. The prevalence of dyslipidaemia in patients with positive adipophilin staining was clearly higher than in those with negative labelling (62.3% vs 13.3%). Relative to the dyslipidaemia risk for patients with negative adipophilin expression, the odds for patients with positive adipophilin expression were increased 10-fold (OR: 10.8; p-value<.01). We identified 23 incident cases of dyslipidaemia over a median follow-up period of 91 months among 54 patients with no history of dyslipidaemia. The patients with positive adipophilin expression showed a higher risk of developing dyslipidaemia (HR: 8.9; p-value<.01). CONCLUSIONS Patients with positive adipophilin staining in their GA biopsies were found to be associated with a higher risk for both baseline and incident dyslipidaemia.
Collapse
Affiliation(s)
- Javier Antoñanzas
- Department of Dermatology, University Clinic of Navarra, Pamplona, Spain
| | - Jorge María Núñez-Córdoba
- Research Support Service, Central Clinical Trials Unit, University Clinic of Navarra, Pamplona, Spain
| | | | | | - Ramón Robledano
- Department of Pathology, University Clinic of Navarra, Pamplona, Spain
| | - Agustín España
- Department of Dermatology, University Clinic of Navarra, Pamplona, Spain.
| |
Collapse
|
15
|
Tran DB, Le NKN, Duong MT, Yuna K, Pham LAT, Nguyen QCT, Tragoolpua Y, Kaewkod T, Kamei K. Drosophila models of the anti-inflammatory and anti-obesity mechanisms of kombucha tea produced by Camellia sinensis leaf fermentation. Food Sci Nutr 2024; 12:5722-5733. [PMID: 39139927 PMCID: PMC11317715 DOI: 10.1002/fsn3.4223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 08/15/2024] Open
Abstract
Kombucha tea is a traditional beverage originating from China and has recently gained popularity worldwide. Kombucha tea is produced by the fermentation of tea leaves and is characterized by its beneficial properties and varied chemical content produced during the fermentation process, which includes organic acids, amino acids, vitamins, minerals, and other biologically active compounds. Kombucha tea is often consumed as a health drink to combat obesity and inflammation; however, the bioactive effects of kombucha tea have not been thoroughly researched. In this study, we reveal the underlying mechanisms of the beneficial properties of kombucha tea and how they protect against obesity and inflammation by studying Drosophila models. We established an inflammatory Drosophila model by knocking down the lipid storage droplet-1 gene, a human perilipin-1 ortholog. In this model, dysfunction of lipid storage droplet-1 induces inflammation by enhancing the infiltration of hemocytes into adipose tissues, increasing reactive oxygen species production, elevating levels of proinflammatory cytokines, and promoting the differentiation of hemocytes into macrophages. These processes are regulated by the c-Jun N-terminal Kinase (JNK) pathway. Using this unique Drosophila model that mimics mammalian inflammation, we verified the beneficial effects of kombucha tea on reducing tissue inflammation. Our data confirms that kombucha tea effectively improves inflammatory conditions by suppressing the expression of cytokines and proinflammatory responses induced by lipid storage droplet-1 dysfunction. It was found that kombucha tea consumption alleviated the production of reactive oxygen species and activated the JNK signaling pathway, signifying its potential as an anti-inflammatory agent against systemic inflammatory responses connected to the JNK pathway. Kombucha tea reduced triglyceride accumulation by increasing the activity of Brummer (a lipase), thereby promoting lipolysis in third-instar larvae. Therefore, kombucha tea could be developed as a novel, functional beverage to protect against obesity and inflammation. Our study also highlights the potential use of this innovative model to evaluate the effects of bioactive compounds derived from natural products.
Collapse
Affiliation(s)
- Duy Binh Tran
- Department of Functional ChemistryKyoto Institute of TechnologyKyotoJapan
- Department of Surgery, College of MedicineUniversity of IllinoisChicagoIllinoisUSA
| | | | - Minh Tue Duong
- Department of Functional ChemistryKyoto Institute of TechnologyKyotoJapan
| | - Kamo Yuna
- Department of Functional ChemistryKyoto Institute of TechnologyKyotoJapan
| | - L. A. Tuan Pham
- Department of Functional ChemistryKyoto Institute of TechnologyKyotoJapan
- Department of Molecular PathologyHanoi Medical UniversityHanoiVietnam
| | - Q. C. Thanh Nguyen
- Department of Functional ChemistryKyoto Institute of TechnologyKyotoJapan
- Department of Chemistry, College of Natural SciencesCantho UniversityCantho CityVietnam
| | - Yingmanee Tragoolpua
- Department of Biology, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Natural Extracts and Innovative Products for Alternative Healthcare Research Group, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Research Center of Deep Technology in Beekeeping and bee Products for Sustainable Development Goals (SMART BEE SDGs), Faculty of ScienceChiang Mai UniversityChiang MaiThailand
| | - Thida Kaewkod
- Department of Biology, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Natural Extracts and Innovative Products for Alternative Healthcare Research Group, Faculty of ScienceChiang Mai UniversityChiang MaiThailand
- Research Center of Deep Technology in Beekeeping and bee Products for Sustainable Development Goals (SMART BEE SDGs), Faculty of ScienceChiang Mai UniversityChiang MaiThailand
| | - Kaeko Kamei
- Department of Functional ChemistryKyoto Institute of TechnologyKyotoJapan
| |
Collapse
|
16
|
Nguyen YDH, Pham TLA, Nishihara T, Kamei K, Tran DB. Depletion of lipid storage droplet-1 delays endoreplication progression and induces cell death in Drosophila salivary gland. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2024; 116:e22132. [PMID: 38993002 DOI: 10.1002/arch.22132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/15/2024] [Accepted: 06/29/2024] [Indexed: 07/13/2024]
Abstract
Perilipins are evolutionarily conserved from insects to mammals. Drosophila lipid storage droplet-1 (LSD-1) is a lipid storage droplet membrane surface-binding protein family member and a counterpart to mammalian perilipin 1 and is known to play a role in lipolysis. However, the function of LSD-1 during specific tissue development remains under investigation. This study demonstrated the role of LSD-1 in salivary gland development. Knockdown of Lsd-1 in the salivary gland was established using the GAL4/UAS system. The third-instar larvae of knockdown flies had small salivary glands containing cells with smaller nuclei. The null mutant Drosophila also showed the same phenotype. The depletion of LSD-1 expression induced a delay of endoreplication due to decreasing CycE expression and increasing DNA damage. Lsd-1 genetically interacted with Myc in the third-instar larvae. These results demonstrate that LSD-1 is involved in cell cycle and cell death programs in the salivary gland, providing novel insight into the effects of LSD-1 in regulating salivary gland development and the interaction between LSD-1 and Myc.
Collapse
Affiliation(s)
- Yen D H Nguyen
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto, Japan
| | - Tuan L A Pham
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto, Japan
| | - Taisei Nishihara
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto, Japan
| | - Kaeko Kamei
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto, Japan
| | - Duy Binh Tran
- Department of Functional Chemistry, Kyoto Institute of Technology, Kyoto, Japan
| |
Collapse
|
17
|
Koenig AB, Tan A, Abdelaal H, Monge F, Younossi ZM, Goodman ZD. Review article: Hepatic steatosis and its associations with acute and chronic liver diseases. Aliment Pharmacol Ther 2024; 60:167-200. [PMID: 38845486 DOI: 10.1111/apt.18059] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/23/2024] [Accepted: 05/13/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Hepatic steatosis is a common finding in liver histopathology and the hallmark of metabolic dysfunction-associated steatotic liver disease (MASLD), formerly known as non-alcoholic fatty liver disease (NAFLD), whose global prevalence is rising. AIMS To review the histopathology of hepatic steatosis and its mechanisms of development and to identify common and rare disease associations. METHODS We reviewed literature on the basic science of lipid droplet (LD) biology and clinical research on acute and chronic liver diseases associated with hepatic steatosis using the PubMed database. RESULTS A variety of genetic and environmental factors contribute to the development of chronic hepatic steatosis or steatotic liver disease, which typically appears macrovesicular. Microvesicular steatosis is associated with acute mitochondrial dysfunction and liver failure. Fat metabolic processes in hepatocytes whose dysregulation leads to the development of steatosis include secretion of lipoprotein particles, uptake of remnant lipoprotein particles or free fatty acids from blood, de novo lipogenesis, oxidation of fatty acids, lipolysis and lipophagy. Hepatic insulin resistance is a key feature of MASLD. Seipin is a polyfunctional protein that facilitates LD biogenesis. Assembly of hepatitis C virus takes place on LD surfaces. LDs make important, functional contact with the endoplasmic reticulum and other organelles. CONCLUSIONS Diverse liver pathologies are associated with hepatic steatosis, with MASLD being the most important contributor. The biogenesis and dynamics of LDs in hepatocytes are complex and warrant further investigation. Organellar interfaces permit co-regulation of lipid metabolism to match generation of potentially toxic lipid species with their LD depot storage.
Collapse
Affiliation(s)
- Aaron B Koenig
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
| | - Albert Tan
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Hala Abdelaal
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Fanny Monge
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| | - Zobair M Younossi
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- The Global NASH Council, Center for Outcomes Research in Liver Diseases, Washington, DC, USA
| | - Zachary D Goodman
- Beatty Liver and Obesity Research Program, Inova Health System, Falls Church, Virginia, USA
- Center for Liver Diseases, Inova Fairfax Hospital, Falls Church, Virginia, USA
| |
Collapse
|
18
|
Mao TH, Huang HQ, Zhang CH. Clinical characteristics and treatment compounds of obesity-related kidney injury. World J Diabetes 2024; 15:1091-1110. [PMID: 38983811 PMCID: PMC11229974 DOI: 10.4239/wjd.v15.i6.1091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 12/22/2023] [Accepted: 04/08/2024] [Indexed: 06/11/2024] Open
Abstract
Disorders in energy homeostasis can lead to various metabolic diseases, particularly obesity. The obesity epidemic has led to an increased incidence of obesity-related nephropathy (ORN), a distinct entity characterized by proteinuria, glomerulomegaly, progressive glomerulosclerosis, and renal function decline. Obesity and its associated renal damage are common in clinical practice, and their incidence is increasing and attracting great attention. There is a great need to identify safe and effective therapeutic modalities, and therapeutics using chemical compounds and natural products are receiving increasing attention. However, the summary is lacking about the specific effects and mechanisms of action of compounds in the treatment of ORN. In this review, we summarize the important clinical features and compound treatment strategies for obesity and obesity-induced kidney injury. We also summarize the pathologic and clinical features of ORN as well as its pathogenesis and potential therapeutics targeting renal inflammation, oxidative stress, insulin resistance, fibrosis, kidney lipid accumulation, and dysregulated autophagy. In addition, detailed information on natural and synthetic compounds used for the treatment of obesity-related kidney disease is summarized. The synthesis of detailed information aims to contribute to a deeper understanding of the clinical treatment modalities for obesity-related kidney diseases, fostering the anticipation of novel insights in this domain.
Collapse
Affiliation(s)
- Tuo-Hua Mao
- Department of Endocrinology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Han-Qi Huang
- Department of Endocrinology, Hubei No. 3 People’s Hospital of Jianghan University, Wuhan 430033, Hubei Province, China
| | - Chuan-Hai Zhang
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390, United States
| |
Collapse
|
19
|
Proença AB, Medeiros GR, Reis GDS, Losito LDF, Ferraz LM, Bargut TCL, Soares NP, Alexandre-Santos B, Campagnole-Santos MJ, Magliano DC, Nobrega ACLD, Santos RAS, Frantz EDC. Adipose tissue plasticity mediated by the counterregulatory axis of the renin-angiotensin system: Role of Mas and MrgD receptors. J Cell Physiol 2024; 239:e31265. [PMID: 38577921 DOI: 10.1002/jcp.31265] [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: 12/07/2023] [Revised: 02/28/2024] [Accepted: 03/18/2024] [Indexed: 04/06/2024]
Abstract
The renin-angiotensin system (RAS) is an endocrine system composed of two main axes: the classical and the counterregulatory, very often displaying opposing effects. The classical axis, primarily mediated by angiotensin receptors type 1 (AT1R), is linked to obesity-associated metabolic effects. On the other hand, the counterregulatory axis appears to exert antiobesity effects through the activation of two receptors, the G protein-coupled receptor (MasR) and Mas-related receptor type D (MrgD). The local RAS in adipose organ has prompted extensive research into white adipose tissue and brown adipose tissue (BAT), with a key role in regulating the cellular and metabolic plasticity of these tissues. The MasR activation favors the brown plasticity signature in the adipose organ by improve the thermogenesis, adipogenesis, and lipolysis, decrease the inflammatory state, and overall energy homeostasis. The MrgD metabolic effects are related to the maintenance of BAT functionality, but the signaling remains unexplored. This review provides a summary of RAS counterregulatory actions triggered by Mas and MrgD receptors on adipose tissue plasticity. Focus on the effects related to the morphology and function of adipose tissue, especially from animal studies, will be given targeting new avenues for treatment of obesity-associated metabolic effects.
Collapse
Affiliation(s)
- Ana Beatriz Proença
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Gabriela Rodrigues Medeiros
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Guilherme Dos Santos Reis
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Luiza da França Losito
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Luiza Mazzali Ferraz
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Thereza Cristina Lonzetti Bargut
- Department of Basic Sciences, Nova Friburgo Health Institute, Fluminense Federal University, Nova Friburgo, Rio de Janeiro, Brazil
| | - Nícia Pedreira Soares
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Beatriz Alexandre-Santos
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Maria Jose Campagnole-Santos
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - D'Angelo Carlo Magliano
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Antonio Claudio Lucas da Nobrega
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and Biophysics, National Institute of Science and Technology in Nanobiopharmaceutics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Eliete Dalla Corte Frantz
- Department of Physiology, Laboratory of Exercise Sciences, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
- Department of Morphology, Research Center on Morphology and Metabolism, Biomedical Institute, Fluminense Federal University, Niteroi, Rio de Janeiro, Brazil
| |
Collapse
|
20
|
Liu X, Li D, Gao W, Chen P, Liu H, Zhao Y, Zhao W, Dong G. Molecular characterization, clinical value, and cancer-immune interactions of genes related to disulfidptosis and ferroptosis in colorectal cancer. Discov Oncol 2024; 15:183. [PMID: 38787520 PMCID: PMC11126553 DOI: 10.1007/s12672-024-01031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND This research strived to construct a new signature utilizing disulfidptosis-related ferroptosis (SRF) genes to anticipate response to immunotherapy, prognosis, and drug sensitivity in individuals with colorectal cancer (CRC). METHODS The data for RNA sequencing as well as corresponding clinical information of individuals with CRC, were extracted from The Cancer Genome Atlas (TCGA) dataset. SRF were constructed with the help of the random forest (RF), least absolute shrinkage and selection operator (LASSO), and stepwise regression algorithms. To validate the SRF model, we applied it to an external cohort, GSE38832. Prognosis, immunotherapy response, drug sensitivity, molecular functions of genes, and somatic mutations of genes were compared across the high- and low-risk groups (categories). Following this, all statistical analyses were conducted with the aid of the R (version 4.23) software and various packages of the Cytoscape (version 3.8.0) tool. RESULTS SRF was developed based on five genes (ATG7, USP7, MMD, PLIN4, and THDC2). Both univariate and multivariate Cox regression analyses established SRF as an independent, prognosis-related risk factor. Individuals from the high-risk category had a more unfavorable prognosis, elevated tumor mutational burden (TMB), and significant immunosuppressive status. Hence, they might have better outcomes post-immunotherapy and might benefit from the administration of pazopanib, lapatinib, and sunitinib. CONCLUSION In conclusion, SRF can act as a new biomarker for prognosis assessment. Moreover, it is also a good predictor of drug sensitivity and immunotherapy response in CRC but should undergo optimization before implementation in clinical settings.
Collapse
Affiliation(s)
- Xianqiang Liu
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Dingchang Li
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wenxing Gao
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Peng Chen
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hao Liu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yingjie Zhao
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wen Zhao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
- School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Guanglong Dong
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
21
|
Majchrzak M, Stojanović O, Ajjaji D, Ben M'barek K, Omrane M, Thiam AR, Klemm RW. Perilipin membrane integration determines lipid droplet heterogeneity in differentiating adipocytes. Cell Rep 2024; 43:114093. [PMID: 38602875 DOI: 10.1016/j.celrep.2024.114093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/12/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
The storage of fat within lipid droplets (LDs) of adipocytes is critical for whole-body health. Acute fatty acid (FA) uptake by differentiating adipocytes leads to the formation of at least two LD classes marked by distinct perilipins (PLINs). How this LD heterogeneity arises is an important yet unresolved cell biological problem. Here, we show that an unconventional integral membrane segment (iMS) targets the adipocyte specific LD surface factor PLIN1 to the endoplasmic reticulum (ER) and facilitates high-affinity binding to the first LD class. The other PLINs remain largely excluded from these LDs until FA influx recruits them to a second LD population. Preventing ER targeting turns PLIN1 into a soluble, cytoplasmic LD protein, reduces its LD affinity, and switches its LD class specificity. Conversely, moving the iMS to PLIN2 leads to ER insertion and formation of a separate LD class. Our results shed light on how differences in organelle targeting and disparities in lipid affinity of LD surface factors contribute to formation of LD heterogeneity.
Collapse
Affiliation(s)
- Mario Majchrzak
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland
| | - Ozren Stojanović
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK
| | - Dalila Ajjaji
- Laboratoire de Physique de l'École Normale Supérieure (ENS), Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005 Paris, France
| | - Kalthoum Ben M'barek
- Laboratoire de Physique de l'École Normale Supérieure (ENS), Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005 Paris, France
| | - Mohyeddine Omrane
- Laboratoire de Physique de l'École Normale Supérieure (ENS), Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005 Paris, France
| | - Abdou Rachid Thiam
- Laboratoire de Physique de l'École Normale Supérieure (ENS), Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005 Paris, France
| | - Robin W Klemm
- Department of Physiology, Anatomy, and Genetics, University of Oxford, Oxford OX1 3PT, UK; Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland.
| |
Collapse
|
22
|
Mazzarella R, Cañón-Beltrán K, Cajas YN, Hamdi M, González EM, da Silveira JC, Leal CLV, Rizos D. Extracellular vesicles-coupled miRNAs from oviduct and uterus modulate signaling pathways related to lipid metabolism and bovine early embryo development. J Anim Sci Biotechnol 2024; 15:51. [PMID: 38570884 PMCID: PMC10993494 DOI: 10.1186/s40104-024-01008-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 02/03/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Extracellular vesicles (EVs) present in oviductal (OF) and uterine fluid (UF) have been shown to enhance bovine embryo quality during in vitro culture by reducing lipid contents and modulating lipid metabolism-related genes (LMGs), while also influencing cell proliferation, suggesting their involvement on the regulation of different biological pathways. The regulation of signaling pathways related to cell differentiation, proliferation, and metabolism is crucial for early embryo development and can determine the success or failure of the pregnancy. Bioactive molecules within EVs in maternal reproductive fluids, such as microRNAs (miRNAs), may contribute to this regulatory process as they modulate gene expression through post-transcriptional mechanisms. RESULTS From the 20 differentially expressed miRNAs, 19 up-regulated in UF-EVs (bta-miR-134, bta-miR-151-3p, bta-miR-155, bta-miR-188, bta-miR-181b, bta-miR-181d, bta-miR-224, bta-miR-23b-3p, bta-miR-24-3p, bta-miR-27a-3p, bta-miR-29a, bta-miR-324, bta-miR-326, bta-miR-345-3p, bta-miR-410, bta-miR-652, bta-miR-677, bta-miR-873 and bta-miR-708) and one (bta-miR-148b) in OF-EVs. These miRNAs were predicted to modulate several pathways such as Wnt, Hippo, MAPK, and lipid metabolism and degradation. Differences in miRNAs found in OF-EVs from the early luteal phase and UF-EVs from mid-luteal phase may reflect different environments to meet the changing needs of the embryo. Additionally, miRNAs may be involved, particularly in the uterus, in the regulation of embryo lipid metabolism, immune system, and implantation. This study evaluated miRNA cargo in OF-EVs from the early luteal phase and UF-EVs from the mid-luteal phase, coinciding with embryo transit within oviduct and uterus in vivo, and its possible influence on LMGs and signaling pathways crucial for early embryo development. A total of 333 miRNAs were detected, with 11 exclusive to OF, 59 to UF, and 263 were common between both groups. CONCLUSIONS Our study suggests that miRNAs within OF- and UF-EVs could modulate bovine embryo development and quality, providing insights into the intricate maternal-embryonic communication that might be involved in modulating lipid metabolism, immune response, and implantation during early pregnancy.
Collapse
Affiliation(s)
| | - Karina Cañón-Beltrán
- Department of Biochemistry and Molecular Biology, Veterinary Faculty, Complutense University of Madrid (UCM), Madrid, Spain
| | - Yulia N Cajas
- Department Agrarian Production, Technical University of Madrid, UPM, Madrid, Spain
- Departamento de Ciencias Biológicas, Universidad Técnica Particular de Loja,, UTPL, Loja, Ecuador
| | - Meriem Hamdi
- Department of Animal Reproduction, INIA-CSIC, Madrid, Spain
| | | | | | - Claudia L V Leal
- Department of Animal Reproduction, INIA-CSIC, Madrid, Spain
- Department of Veterinary Medicine, FZEA-USP, Pirassununga, Brazil
| | - D Rizos
- Department of Animal Reproduction, INIA-CSIC, Madrid, Spain.
| |
Collapse
|
23
|
Hsia JZ, Liu D, Haynes L, Cruz-Cosme R, Tang Q. Lipid Droplets: Formation, Degradation, and Their Role in Cellular Responses to Flavivirus Infections. Microorganisms 2024; 12:647. [PMID: 38674592 PMCID: PMC11051834 DOI: 10.3390/microorganisms12040647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 03/18/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Lipid droplets (LDs) are cellular organelles derived from the endoplasmic reticulum (ER), serving as lipid storage sites crucial for maintaining cellular lipid homeostasis. Recent attention has been drawn to their roles in viral replication and their interactions with viruses. However, the precise biological functions of LDs in viral replication and pathogenesis remain incompletely understood. To elucidate the interaction between LDs and viruses, it is imperative to comprehend the biogenesis of LDs and their dynamic interactions with other organelles. In this review, we explore the intricate pathways involved in LD biogenies within the cytoplasm, encompassing the uptake of fatty acid from nutrients facilitated by CD36-mediated membranous protein (FABP/FATP)-FA complexes, and FA synthesis via glycolysis in the cytoplasm and the TCL cycle in mitochondria. While LD biogenesis primarily occurs in the ER, matured LDs are intricately linked to multiple organelles. Viral infections can lead to diverse consequences in terms of LD status within cells post-infection, potentially involving the breakdown of LDs through the activation of lipophagy. However, the exact mechanisms underlying LD destruction or accumulation by viruses remain elusive. The significance of LDs in viral replication renders them effective targets for developing broad-spectrum antivirals. Moreover, considering that reducing neutral lipids in LDs is a strategy for anti-obesity treatment, LD depletion may not pose harm to cells. This presents LDs as promising antiviral targets for developing therapeutics that are minimally or non-toxic to the host.
Collapse
Affiliation(s)
| | | | | | | | - Qiyi Tang
- Department of Microbiology, Howard University College of Medicine, Washington, DC 20059, USA; (J.Z.H.); (D.L.); (L.H.); (R.C.-C.)
| |
Collapse
|
24
|
Desgrouas C, Thalheim T, Cerino M, Badens C, Bonello-Palot N. Perilipin 1: a systematic review on its functions on lipid metabolism and atherosclerosis in mice and humans. Cardiovasc Res 2024; 120:237-248. [PMID: 38214891 DOI: 10.1093/cvr/cvae005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 10/12/2023] [Accepted: 10/27/2023] [Indexed: 01/13/2024] Open
Abstract
The function of perilipin 1 in human metabolism was recently highlighted by the description of PLIN1 variants associated with various pathologies. These include severe familial partial lipodystrophy and early onset acute coronary syndrome. Additionally, certain variants have been reported to have a protective effect on cardiovascular diseases. The role of this protein remains controversial in mice and variant interpretation in humans is still conflicting. This literature review has two primary objectives (i) to clarify the function of the PLIN1 gene in lipid metabolism and atherosclerosis by examining functional studies performed in cells (adipocytes) and mice and (ii) to understand the impact of PLIN1 variants identified in humans based on the variant's location within the protein and the type of variant (missense or frameshift). To achieve these objectives, we conducted an extensive analysis of the relevant literature on perilipin 1, its function in cellular models and mice, and the consequences of its mutations in humans. We also utilized bioinformatics tools and consulted the Human Genetics Cardiovascular Disease Knowledge Portal to enhance the pathogenicity assessment of PLIN1 missense variants.
Collapse
Affiliation(s)
- Camille Desgrouas
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Faculte de médecine, 27 Bd Jean Moulin 13005 Marseille, France
| | - Tabea Thalheim
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Faculte de médecine, 27 Bd Jean Moulin 13005 Marseille, France
| | - Mathieu Cerino
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Faculte de médecine, 27 Bd Jean Moulin 13005 Marseille, France
- AP-HM, Service de Biochimie, Hôpital de la Timone 264 rue Saint Pierre 13005 Marseille, France
| | - Catherine Badens
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Faculte de médecine, 27 Bd Jean Moulin 13005 Marseille, France
- AP-HM, Service de Biochimie, Hôpital de la Timone 264 rue Saint Pierre 13005 Marseille, France
- Département de Génétique Médicale, APHM, Hôpital Timone Enfants, Hôpital de la Timone 264 rue Saint Pierre 13005 Marseille, France
| | - Nathalie Bonello-Palot
- Aix Marseille Univ, INSERM, Marseille Medical Genetics, Faculte de médecine, 27 Bd Jean Moulin 13005 Marseille, France
- Département de Génétique Médicale, APHM, Hôpital Timone Enfants, Hôpital de la Timone 264 rue Saint Pierre 13005 Marseille, France
| |
Collapse
|
25
|
Cinato M, Andersson L, Miljanovic A, Laudette M, Kunduzova O, Borén J, Levin MC. Role of Perilipins in Oxidative Stress-Implications for Cardiovascular Disease. Antioxidants (Basel) 2024; 13:209. [PMID: 38397807 PMCID: PMC10886189 DOI: 10.3390/antiox13020209] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Oxidative stress is the imbalance between the production of reactive oxygen species (ROS) and antioxidants in a cell. In the heart, oxidative stress may deteriorate calcium handling, cause arrhythmia, and enhance maladaptive cardiac remodeling by the induction of hypertrophic and apoptotic signaling pathways. Consequently, dysregulated ROS production and oxidative stress have been implicated in numerous cardiac diseases, including heart failure, cardiac ischemia-reperfusion injury, cardiac hypertrophy, and diabetic cardiomyopathy. Lipid droplets (LDs) are conserved intracellular organelles that enable the safe and stable storage of neutral lipids within the cytosol. LDs are coated with proteins, perilipins (Plins) being one of the most abundant. In this review, we will discuss the interplay between oxidative stress and Plins. Indeed, LDs and Plins are increasingly being recognized for playing a critical role beyond energy metabolism and lipid handling. Numerous reports suggest that an essential purpose of LD biogenesis is to alleviate cellular stress, such as oxidative stress. Given the yet unmet suitability of ROS as targets for the intervention of cardiovascular disease, the endogenous antioxidant capacity of Plins may be beneficial.
Collapse
Affiliation(s)
- Mathieu Cinato
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Linda Andersson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Azra Miljanovic
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Marion Laudette
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University—Paul Sabatier, 31432 Toulouse, France;
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Malin C. Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| |
Collapse
|
26
|
Kulik U, Moesta C, Spanel R, Borlak J. Dysfunctional Cori and Krebs cycle and inhibition of lactate transporters constitute a mechanism of primary nonfunction of fatty liver allografts. Transl Res 2024; 264:33-65. [PMID: 37722450 DOI: 10.1016/j.trsl.2023.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/06/2023] [Accepted: 09/06/2023] [Indexed: 09/20/2023]
Abstract
Orthotopic liver transplantation (OLT) is a lifesaving procedure. However, grafts may fail due to primary nonfunction (PNF). In the past, we demonstrated PNFs to be mainly associated with fatty allografts, and given its unpredictable nature, the development of a disease model is urgently needed. In an effort to investigate mechanism of fatty allograft-associated PNFs, we induced fatty liver disease in donor animals by feeding rats a diet deficient in methionine and choline (MCD). We performed OLT with allografts of different grades of hepatic steatosis and compared the results to healthy ones. We assessed liver function by considering serum biochemistries, and investigated genome wide responses following OLT of healthy and fatty allograft-associated PNFs. Furthermore, we performed immunohistochemistry to evaluate markers of oxidative stress and reperfusion injury, inflammation, glycolysis and gluconeogenesis, lactate transport, and its utilization as part of the Cori cycle. Strikingly, PNFs are strictly lipid content dependent. Nonetheless, a fat content of ≤17% and an increase in the size of hepatocytes of ≤11% (ballooning) greatly improved outcome of OLTs and the hepatic microcirculation. Mechanistically, PNFs arise from a dysfunctional Cori cycle with complete ablation of the lactate transporter SLC16A1. Thus, lipid-laden hepatocytes fail to perform gluconeogenesis via lactate reutilization, and the resultant hyperlactatemia and lactic acidosis causes cardiac arrhythmogenicity and death. Furthermore, the genomic and immunohistochemistry investigations underscore a dysfunctional Krebs cycle with impaired energy metabolism in lipid-burdened mitochondria. Together, we show fatty allografts to be highly vulnerable towards ischemia/reperfusion-injury, and stabilizing the Cori cycle is of critical importance to avert PNFs.
Collapse
Affiliation(s)
- Ulf Kulik
- Department of General, Visceral- and Transplantation Surgery, Hannover Medical School, Hannover, Germany
| | - Caroline Moesta
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Reinhard Spanel
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
27
|
Martin Carli JF, Dzieciatkowska M, Hernandez TL, Monks J, McManaman JL. Comparative proteomic analysis of human milk fat globules and paired membranes and mouse milk fat globules identifies core cellular systems contributing to mammary lipid trafficking and secretion. Front Mol Biosci 2023; 10:1259047. [PMID: 38169886 PMCID: PMC10759240 DOI: 10.3389/fmolb.2023.1259047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction: Human milk delivers critical nutritional and immunological support to human infants. Milk fat globules (MFGs) and their associated membranes (MFGMs) contain the majority of milk lipids and many bioactive components that contribute to neonatal development and health, yet their compositions have not been fully defined, and the mechanisms responsible for formation of these structures remain incompletely understood. Methods: In this study, we used untargeted mass spectrometry to quantitatively profile the protein compositions of freshly obtained MFGs and their paired, physically separated MFGM fractions from 13 human milk samples. We also quantitatively profiled the MFG protein compositions of 9 pooled milk samples from 18 lactating mouse dams. Results: We identified 2,453 proteins and 2,795 proteins in the majority of human MFG and MFGM samples, respectively, and 1,577 proteins in mouse MFGs. Using paired analyses of protein abundance in MFGMs compared to MFGs (MFGM-MFG; 1% FDR), we identified 699 proteins that were more highly abundant in MFGMs (MFGM-enriched), and 201 proteins that were less abundant in MFGMs (cytoplasmic). MFGM-enriched proteins comprised membrane systems (apical plasma membrane and multiple vesicular membranes) hypothesized to be responsible for lipid and protein secretion and components of membrane transport and signaling systems. Cytoplasmic proteins included ribosomal and proteasomal systems. Comparing abundance between human and mouse MFGs, we found a positive correlation (R 2 = 0.44, p < 0.0001) in the relative abundances of 1,279 proteins that were found in common across species. Discussion: Comparative pathway enrichment analyses between human and mouse samples reveal similarities in membrane trafficking and signaling pathways involved in milk fat secretion and identify potentially novel immunological components of MFGs. Our results advance knowledge of the composition and relative quantities of proteins in human and mouse MFGs in greater detail, provide a quantitative profile of specifically enriched human MFGM proteins, and identify core cellular systems involved in milk lipid secretion.
Collapse
Affiliation(s)
- Jayne F. Martin Carli
- Section of Nutrition, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Teri L. Hernandez
- College of Nursing, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jenifer Monks
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - James L. McManaman
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
28
|
Stribny J, Schneiter R. Binding of perilipin 3 to membranes containing diacylglycerol is mediated by conserved residues within its PAT domain. J Biol Chem 2023; 299:105384. [PMID: 37898398 PMCID: PMC10694602 DOI: 10.1016/j.jbc.2023.105384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 10/12/2023] [Accepted: 10/17/2023] [Indexed: 10/30/2023] Open
Abstract
Perilipins (PLINs) constitute an evolutionarily conserved family of proteins that specifically associate with the surface of lipid droplets (LDs). These proteins function in LD biogenesis and lipolysis and help to stabilize the surface of LDs. PLINs are typically composed of three different protein domains. They share an N-terminal PAT domain of unknown structure and function, a central region containing 11-mer repeats that form amphipathic helices, and a C-terminal domain that adopts a 4-helix bundle structure. How exactly these three distinct domains contribute to PLIN function remains to be determined. Here, we show that the N-terminal PAT domain of PLIN3 binds diacylglycerol (DAG), the precursor to triacylglycerol, a major storage lipid of LDs. PLIN3 and its PAT domain alone bind liposomes with micromolar affinity and PLIN3 binds artificial LDs containing low concentrations of DAG with nanomolar affinity. The PAT domain of PLIN3 is predicted to adopt an amphipathic triangular shaped structure. In silico ligand docking indicates that DAG binds to one of the highly curved regions within this domain. A conserved aspartic acid residue in the PAT domain, E86, is predicted to interact with DAG, and we found that its substitution abrogates high affinity binding of DAG as well as DAG-stimulated association with liposome and artificial LDs. These results indicate that the PAT domain of PLINs harbor specific lipid-binding properties that are important for targeting these proteins to the surface of LDs and to ER membrane domains enriched in DAG to promote LD formation.
Collapse
Affiliation(s)
- Jiri Stribny
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
29
|
Pan J, Jin Y, Jin H, Li C, Zhang Y, Liu Y, Jin G, Zhao J, He L, Sheng L. New insights into the function of lipid droplet-related proteins and lipid metabolism of salt-stimulated porcine biceps femoris: label-free quantitative phosphoproteomics, morphometry and bioinformatics. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:7517-7528. [PMID: 37440710 DOI: 10.1002/jsfa.12857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/22/2023] [Accepted: 07/14/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Lipid droplets (LDs) are important multifunctional organelles responsible for lipid metabolism of postmortem muscle. However, the dynamics in their building blocks (cores and layers) and phosphorylation of lipid droplet-related proteins (LDRPs) regulating meat lipolysis remain unknown at salt-stimulated conditions. RESULTS LDRPs extracted from cured porcine biceps femoris (1% and 3% salt) were subjected to label-free quantitative phosphoproteomic analysis and LDs morphological validation. Results indicated that 3% salt curing significantly decreased triglyceride (TG) content with increase in glycerol and decrease in LDs fluorescence compared to 1% salt curing. Comparative phosphoproteomics showed that there were significant changes in phosphorylation at 386 sites on 174 LDRPs between assayed groups (P < 0.05). These differential proteins were mainly involved in lipid and carbohydrate metabolism. Curing of 3% salt induced more site-specific phosphorylation of perilipin 1 (PLIN1, at Ser81) and adipose triglyceride lipase (ATGL, at Ser399) than 1%, whereas the phosphorylation (at Ser600) of hormone-sensitive lipase (HSL) was up-regulated. Ultrastructure imaging showed that LDs were mostly associated with mitochondria, and the average diameter of LDs decreased from 2.34 μm (1% salt) to 1.73 μm (3% salt). CONCLUSION Phosphoproteomics unraveled salt-stimulated LDRPs phosphorylation of cured porcine meat provoked intensified lipolysis. Curing of 3% salt allowed an enhanced lipolysis than 1% by up-regulating the phosphorylation sites of LDRPs and recruited lipases. The visible splitting of LDs, together with sarcoplasmic disorganization, supported the lipolysis robustness following 3% salt curing. The finding provides optimization ideas for high-quality production of cured meat products. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiajing Pan
- School of Food and Health, Beijing Technology and Business University, Beijing, China
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yongguo Jin
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Haobo Jin
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Chengliang Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yan Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Yuanyi Liu
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Guofeng Jin
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Jianying Zhao
- College of Tea and Food Science Technology, Jiangsu Vocational College Agriculture and Forestry, Jurong, China
| | - Lichao He
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Long Sheng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
30
|
Kumari RM, Khatri A, Chaudhary R, Choudhary V. Concept of lipid droplet biogenesis. Eur J Cell Biol 2023; 102:151362. [PMID: 37742390 PMCID: PMC7615795 DOI: 10.1016/j.ejcb.2023.151362] [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] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 09/26/2023] Open
Abstract
Lipid droplets (LD) are functionally conserved fat storage organelles found in all cell types. LDs have a unique structure comprising of a hydrophobic core of neutral lipids (fat), triacylglycerol (TAG) and cholesterol esters (CE) surrounded by a phospholipid monolayer. LD surface is decorated by a multitude of proteins and enzymes rendering this compartment functional. Accumulating evidence suggests that LDs originate from discrete ER-subdomains, demarcated by the lipodystrophy protein seipin, however, the mechanisms of which are not well understood. LD biogenesis factors together with biophysical properties of the ER membrane orchestrate spatiotemporal regulation of LD nucleation and growth at specific ER subdomains in response to metabolic cues. Defects in LD formation manifests in several human pathologies, including obesity, lipodystrophy, ectopic fat accumulation, and insulin resistance. Here, we review recent advances in understanding the molecular events during initial stages of eukaryotic LD assembly and discuss the critical role of factors that ensure fidelity of this process.
Collapse
Affiliation(s)
- R Mankamna Kumari
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Amit Khatri
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Ritika Chaudhary
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Vineet Choudhary
- Lipid Metabolism Laboratory, Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
31
|
Dargham T, Mallick I, Kremer L, Santucci P, Canaan S. Intrabacterial lipid inclusion-associated proteins: a core machinery conserved from saprophyte Actinobacteria to the human pathogen Mycobacterium tuberculosis. FEBS Open Bio 2023; 13:2306-2323. [PMID: 37872001 PMCID: PMC10699116 DOI: 10.1002/2211-5463.13721] [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: 09/05/2023] [Revised: 10/02/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the aetiologic agent of tuberculosis (TB), stores triacylglycerol (TAG) in the form of intrabacterial lipid inclusions (ILI) to survive and chronically persist within its host. These highly energetic molecules represent a major source of carbon to support bacterial persistence and reactivation, thus playing a leading role in TB pathogenesis. However, despite its physiological and clinical relevance, ILI metabolism in Mtb remains poorly understood. Recent discoveries have suggested that several ILI-associated proteins might be widely conserved across TAG-producing prokaryotes, but still very little is known regarding the nature and the biological functions of these proteins. Herein, we performed an in silico analysis of three independent ILI-associated proteomes previously reported to computationally define a potential core ILI-associated proteome, referred to as ILIome. Our investigation revealed the presence of 70 orthologous proteins that were strictly conserved, thereby defining a minimal ILIome core. We further narrowed our analysis to proteins involved in lipid metabolism and discuss here their putative biological functions, along with their molecular interactions and dynamics at the surface of these bacterial organelles. We also highlight the experimental limitations of the original proteomic investigations and of the present bioinformatic analysis, while describing new technological approaches and presenting biological perspectives in the field. The in silico investigation presented here aims at providing useful datasets that could constitute a scientific resource of broad interest for the mycobacterial community, with the ultimate goal of enlightening ILI metabolism in prokaryotes with a special emphasis on Mtb pathogenesis.
Collapse
Affiliation(s)
- Tonia Dargham
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
- IHU Méditerranée InfectionAix‐Marseille Univ.France
| | - Ivy Mallick
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM)Université de MontpellierFrance
- INSERM, Institut de Recherche en Infectiologie de MontpellierFrance
| | - Pierre Santucci
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
| | - Stéphane Canaan
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
| |
Collapse
|
32
|
Pan H, Qin Y, Zhu J, Wang W, Liu Z, Huang X, Lam SM, Shui G, Wang Y, Jiang Y, Huang X. Centrins control chicken cone cell lipid droplet dynamics through lipid-droplet-localized SPDL1. Dev Cell 2023; 58:2528-2544.e8. [PMID: 37699389 DOI: 10.1016/j.devcel.2023.08.012] [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: 07/30/2022] [Revised: 05/10/2023] [Accepted: 08/08/2023] [Indexed: 09/14/2023]
Abstract
As evolutionarily conserved organelles, lipid droplets (LDs) carry out numerous functions and have various subcellular localizations in different cell types and species. In avian cone cells, there is a single apically localized LD. We demonstrated that CIDEA (cell death inducing DFFA like effector a) and microtubules promote the formation of the single LD in chicken cone cells. Centrins, which are well-known centriole proteins, target to the cone cell LD via their C-terminal calcium-binding domains. Centrins localize on cone cell LDs with the help of SPDL1-L (spindle apparatus coiled-coil protein 1-L), a previously uncharacterized isoform of the kinetochore-associated dynein adaptor SPDL1. The loss of CETN3 or overexpression of a truncated CETN1 abrogates the apical localization of the cone cell LD. Simulation analysis showed that multiple LDs or a single mispositioned LD reduces the light sensitivity. Collectively, our findings identify a role of centrins in the regulation of cone cell LD localization, which is important for the light sensitivity of cone cells.
Collapse
Affiliation(s)
- Huimin Pan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaqiang Qin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinglin Zhu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhonghua Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sin Man Lam
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guanghou Shui
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuqiang Jiang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450001, China.
| |
Collapse
|
33
|
Zocchi M, Bartolini M, Maier JA, Castiglioni S. Low extracellular magnesium induces phenotypic and metabolic alterations in C2C12-derived myotubes. Sci Rep 2023; 13:19425. [PMID: 37940675 PMCID: PMC10632379 DOI: 10.1038/s41598-023-46543-9] [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: 08/02/2023] [Accepted: 11/02/2023] [Indexed: 11/10/2023] Open
Abstract
Magnesium (Mg) has a pivotal role in upholding skeletal muscle health and optimizing performance. Its deficiency decreases muscle strength, and an association has been reported between Mg intake and sarcopenia. To gain a comprehensive understanding of the repercussions arising from low Mg concentrations on muscle behavior, we employed an in vitro model utilizing C2C12-derived myotubes. Myotubes cultured in low Mg show a significant reduction of thickness and a concomitant down-regulation of myosin heavy chain (MyHC), Myog and Myomixer. In parallel, myotubes shape their metabolism. Glycolysis is inhibited and beta-oxidation increases. These metabolic changes are consistent with the increase of MyHC I (slow) vs. MyHC II (fast) expression. We identified an essential player in these changes, namely nitric oxide (NO), as the increase in NO production appeared to orchestrate the observed modifications in myotube behavior and metabolism under low Mg conditions. Understanding these underlying mechanisms may pave the way for targeted interventions to ameliorate muscle-related conditions associated with Mg deficiency and contribute to enhancing overall muscle health and function.
Collapse
Affiliation(s)
- Monica Zocchi
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Italy
| | - Marco Bartolini
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Italy
| | - Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Italy
| | - Sara Castiglioni
- Department of Biomedical and Clinical Sciences, Università di Milano, 20157, Milano, Italy.
| |
Collapse
|
34
|
Sivaraman K, Pino P, Raussin G, Anchisi S, Metayer C, Dagany N, Held J, Wrenger S, Welte T, Wurm MJ, Wurm FM, Olejnicka B, Janciauskiene S. Human PBMCs Form Lipid Droplets in Response to Spike Proteins. Microorganisms 2023; 11:2683. [PMID: 38004695 PMCID: PMC10672762 DOI: 10.3390/microorganisms11112683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/22/2023] [Accepted: 10/30/2023] [Indexed: 11/26/2023] Open
Abstract
Intracellular lipid droplets (LDs) can accumulate in response to inflammation, metabolic stresses, and other physiological/pathological processes. Herein, we investigated whether spike proteins of SARS-CoV-2 induce LDs in human peripheral blood mononuclear cells (PBMCs) and in pulmonary microvascular endothelial cells (HPMECs). PBMCs or HPMECs were incubated alone or with endotoxin-free recombinant variants of trimeric spike glycoproteins (Alpha, Beta, Delta, and Omicron, 12 µg/mL). Afterward, cells were stained with Oil Red O for LDs, cytokine release was determined through ELISA, and the gene expression was analyzed through real-time PCR using TaqMan assays. Our data show that spikes induce LDs in PBMCs but not in HPMECs. In line with this, in PBMCs, spike proteins lower the expression of genes involving lipid metabolism and LD formation, such as SREBF1, HMGCS1, LDLR, and CD36. On the other hand, PBMCs exposed to spikes for 6 or 18 h did not increase in IL-1β, IL-6, IL-8, MCP-1, and TNFα release or expression as compared to non-treated controls. Thus, spike-induced LD formation in PBMCs seems to not be related to cell inflammatory activation. Further detailed studies are warranted to investigate in which specific immune cells spikes induce LDs, and what are the pathophysiological mechanisms and consequences of this induction in vivo.
Collapse
Affiliation(s)
- Kokilavani Sivaraman
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Paco Pino
- ExcellGene SA, 1970 Monthey, Switzerland
| | | | | | | | | | - Julia Held
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Sabine Wrenger
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | | | - Florian M. Wurm
- ExcellGene SA, 1970 Monthey, Switzerland
- Faculty of Life Sciences, École Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Beata Olejnicka
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
35
|
Subramaiyam N. Insights of mitochondrial involvement in alcoholic fatty liver disease. J Cell Physiol 2023; 238:2175-2190. [PMID: 37642259 DOI: 10.1002/jcp.31100] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/31/2023]
Abstract
Alcoholic liver disease (ALD) is a global concern affecting most of the population and leading to the development of end-stage liver disease. Metabolic alterations due to increased alcohol consumption surge the hepatic accumulation of lipids and develop into a severe form of alcoholic steatohepatitis (ASH), depending on age and the consumption rate. The mitochondria in the hepatocyte actively regulate metabolic homeostasis and are disrupted in ALD pathogenesis. The increased NADH upon ethanol metabolism inhibits the mitochondrial oxidation of fatty acids, alters oxidative phosphorylation, and favors de novo lipogenesis. The higher mitochondrial respiration in early ALD increases free radical generation, whereas mitochondrial respiration is uncoupled in chronic ALD, affecting the cellular energy status. The defective glutathione importer due to excessive cholesterol loading and low adenosine triphosphate accounts for additional oxidative stress leading to hepatocyte apoptosis. The defective mitochondrial transcription machinery and sirtuins function in ALD affect mitochondrial function and biogenesis. The metabolites of ethanol metabolism epigenetically alter the gene expression profile of hepatic cell populations by modulating the promoters and sirtuins, aiding hepatic fibrosis and inflammation. The defect in mitophagy increases the accumulation of megamitochondria in hepatocytes and attracts immune cells by releasing mitochondrial damage-associated molecular patterns to initiate hepatic inflammation and ASH progression. Thus, maintaining mitochondrial lipid homeostasis and antioxidant capacity pharmacologically could provide a better outcome for ALD management.
Collapse
Affiliation(s)
- Nithyananthan Subramaiyam
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
36
|
Zhu J, Zhou Y, Jin B, Shu J. Role of estrogen in the regulation of central and peripheral energy homeostasis: from a menopausal perspective. Ther Adv Endocrinol Metab 2023; 14:20420188231199359. [PMID: 37719789 PMCID: PMC10504839 DOI: 10.1177/20420188231199359] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 08/16/2023] [Indexed: 09/19/2023] Open
Abstract
Estrogen plays a prominent role in regulating and coordinating energy homeostasis throughout the growth, development, reproduction, and aging of women. Estrogen receptors (ERs) are widely expressed in the brain and nearly all tissues of the body. Within the brain, central estrogen via ER regulates appetite and energy expenditure and maintains cell glucose metabolism, including glucose transport, aerobic glycolysis, and mitochondrial function. In the whole body, estrogen has shown beneficial effects on weight control, fat distribution, glucose and insulin resistance, and adipokine secretion. As demonstrated by multiple in vitro and in vivo studies, menopause-related decline of circulating estrogen may induce the disturbance of metabolic signals and a significant decrease in bioenergetics, which could trigger an increased incidence of late-onset Alzheimer's disease, type 2 diabetes mellitus, hypertension, and cardiovascular diseases in postmenopausal women. In this article, we have systematically reviewed the role of estrogen and ERs in body composition and lipid/glucose profile variation occurring with menopause, which may provide a better insight into the efficacy of hormone therapy in maintaining energy metabolic homeostasis and hold a clue for development of novel therapeutic approaches for target tissue diseases.
Collapse
Affiliation(s)
- Jing Zhu
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yier Zhou
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Bihui Jin
- Center for Reproductive Medicine, Department of Reproductive Endocrinology, Zhejiang Provincial People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jing Shu
- Reproductive Medicine Center, the First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310006, China
| |
Collapse
|
37
|
Scrimieri R, Locatelli L, Cazzaniga A, Cazzola R, Malucelli E, Sorrentino A, Iotti S, Maier JA. Ultrastructural features mirror metabolic derangement in human endothelial cells exposed to high glucose. Sci Rep 2023; 13:15133. [PMID: 37704683 PMCID: PMC10499809 DOI: 10.1038/s41598-023-42333-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
High glucose-induced endothelial dysfunction is the early event that initiates diabetes-induced vascular disease. Here we employed Cryo Soft X-ray Tomography to obtain three-dimensional maps of high D-glucose-treated endothelial cells and their controls at nanometric spatial resolution. We then correlated ultrastructural differences with metabolic rewiring. While the total mitochondrial mass does not change, high D-glucose promotes mitochondrial fragmentation, as confirmed by the modulation of fission-fusion markers, and dysfunction, as demonstrated by the drop of membrane potential, the decreased oxygen consumption and the increased production of reactive oxygen species. The 3D ultrastructural analysis also indicates the accumulation of lipid droplets in cells cultured in high D-glucose. Indeed, because of the decrease of fatty acid β-oxidation induced by high D-glucose concentration, triglycerides are esterified into fatty acids and then stored into lipid droplets. We propose that the increase of lipid droplets represents an adaptive mechanism to cope with the overload of glucose and associated oxidative stress and metabolic dysregulation.
Collapse
Affiliation(s)
- Roberta Scrimieri
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy.
| | - Laura Locatelli
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy
| | - Alessandra Cazzaniga
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy
| | - Roberta Cazzola
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy
| | - Emil Malucelli
- Department of Pharmacy and Biotechnology, Università di Bologna, 40127, Bologna, Italy
| | - Andrea Sorrentino
- Mistral Beamline, ALBA Synchrotron Light Source, Cerdanyola del Valles, 08290, Barcelona, Spain
| | - Stefano Iotti
- Department of Pharmacy and Biotechnology, Università di Bologna, 40127, Bologna, Italy
- National Institute of Biostructures and Biosystems, Viale Delle Medaglie d'Oro 305, 00136, Rome, Italy
| | - Jeanette A Maier
- Department of Biomedical and Clinical Sciences, Università degli Studi di Milano, 20157, Milan, Italy.
| |
Collapse
|
38
|
Zhang C. Coherent Raman scattering microscopy of lipid droplets in cells and tissues. JOURNAL OF RAMAN SPECTROSCOPY : JRS 2023; 54:988-1000. [PMID: 38076450 PMCID: PMC10707480 DOI: 10.1002/jrs.6540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/03/2023] [Indexed: 09/03/2024]
Abstract
Lipid droplets (LDs) play a key role as the hub for lipid metabolism to maintain cellular metabolic homeostasis. Understanding the functions and changes of LDs in different pathological conditions is crucial for identifying new markers for diagnosis and discovering new targets for treatment. In recent years, coherent Raman scattering (CRS) microscopy has been popularized for the imaging and quantification of LDs in live cells. Compared to spontaneous Raman scattering microscopy, CRS microscopy offers a much higher imaging speed while maintaining similar chemical information. Due to the high lipid density, LDs usually have strong CRS signals and therefore are the most widely studied organelle in the CRS field. In this review, we discuss recent achievements using CRS to study the quantity, distribution, composition, and dynamics of LDs in various systems.
Collapse
Affiliation(s)
- Chi Zhang
- Department of Chemistry, Purdue Center for Cancer Research, Purdue Institute of Inflammation Immunology and Infectious Disease, Purdue University, West Lafayette, IN
| |
Collapse
|
39
|
Yao D, Ranadheera CS, Shen C, Wei W, Cheong LZ. Milk fat globule membrane: composition, production and its potential as encapsulant for bioactives and probiotics. Crit Rev Food Sci Nutr 2023; 64:12336-12351. [PMID: 37632418 DOI: 10.1080/10408398.2023.2249992] [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] [Indexed: 08/28/2023]
Abstract
Milk fat globule membrane (MFGM) is a complex trilayer structure present in mammalian milk and is mainly composed of phospholipids and proteins (>90%). Many studies revealed MFGM has positive effects on the immune system, brain development, and cognitive function of infants. Probiotics are live microorganisms that have been found to improve mental health and insulin sensitivity, regulate immunity, and prevent allergies. Probiotics are unstable and prone to degradation by environmental, processing, and storage conditions. In this review, the processes used for encapsulation of probiotics particularly the potential of MFGM and its constituents as encapsulating materials for probiotics are described. This study analyzes the importance of MFGM in encapsulating bioactive substances and emphasizes the interaction with probiotics and the gut as well as its resistance to adverse environmental factors in the digestive system when used as a probiotic embedding material. MFGM can enhance the gastric acid resistance and bile resistance of probiotics, mainly manifested in the survival rate of probiotics. Due to the role of digestion, MFGM-coated probiotics can be released in the intestine, and due to the biocompatibility of the membrane, it can promote the binding of probiotics to intestinal epithelial cells, and promote the colonization of some probiotics in the intestine.
Collapse
Affiliation(s)
- Dan Yao
- Zhejiang-Malaysia Joint Research Laboratory for Agricultural Product Processing and Nutrition, College of Food and Pharmaceutical Science, Ningbo University, Ningbo, China
| | - Chaminda Senaka Ranadheera
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, Melbourne, Victoria, Australia
| | - Cai Shen
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, Melbourne, Victoria, Australia
- China Beacons Institute, University of Nottingham Ningbo China, Ningbo, China
| | - Wei Wei
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Ling-Zhi Cheong
- School of Agriculture, Food and Ecosystem Sciences, Faculty of Science, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
40
|
Bombarda-Rocha V, Silva D, Badr-Eddine A, Nogueira P, Gonçalves J, Fresco P. Challenges in Pharmacological Intervention in Perilipins (PLINs) to Modulate Lipid Droplet Dynamics in Obesity and Cancer. Cancers (Basel) 2023; 15:4013. [PMID: 37568828 PMCID: PMC10417315 DOI: 10.3390/cancers15154013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Perilipins (PLINs) are the most abundant proteins in lipid droplets (LD). These LD-associated proteins are responsible for upgrading LD from inert lipid storage structures to fully functional organelles, fundamentally integrated in the lipid metabolism. There are five distinct perilipins (PLIN1-5), each with specific expression patterns and metabolic activation, but all capable of regulating the activity of lipases on LD. This plurality creates a complex orchestrated mechanism that is directly related to the healthy balance between lipogenesis and lipolysis. Given the essential role of PLINs in the modulation of the lipid metabolism, these proteins can become interesting targets for the treatment of lipid-associated diseases. Since reprogrammed lipid metabolism is a recognized cancer hallmark, and obesity is a known risk factor for cancer and other comorbidities, the modulation of PLINs could either improve existing treatments or create new opportunities for the treatment of these diseases. Even though PLINs have not been, so far, directly considered for pharmacological interventions, there are many established drugs that can modulate PLINs activity. Therefore, the aim of this study is to assess the involvement of PLINs in diseases related to lipid metabolism dysregulation and whether PLINs can be viewed as potential therapeutic targets for cancer and obesity.
Collapse
Affiliation(s)
- Victória Bombarda-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Dany Silva
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Allal Badr-Eddine
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
| | - Patrícia Nogueira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Jorge Gonçalves
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Paula Fresco
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (V.B.-R.); (D.S.); (A.B.-E.); (P.N.); (P.F.)
- UCIBIO–Applied Molecular Biosciences Unit, Associate Laboratory i4HB, Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
41
|
Qu Y, Wang W, Xiao MZX, Zheng Y, Liang Q. The interplay between lipid droplets and virus infection. J Med Virol 2023; 95:e28967. [PMID: 37496184 DOI: 10.1002/jmv.28967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/28/2023]
Abstract
As an intracellular parasite, the virus usurps cellular machinery and modulates cellular metabolism pathways to replicate itself in cells. Lipid droplets (LDs) are universally conserved energy storage organelles that not only play vital roles in maintaining lipid homeostasis but are also involved in viral replication. Increasing evidence has demonstrated that viruses take advantage of cellular lipid metabolism by targeting the biogenesis, hydrolysis, and lipophagy of LD during viral infection. In this review, we summarize the current knowledge about the modulation of cellular LD by different viruses, with a special emphasis on the Hepatitis C virus, Dengue virus, and SARS-CoV-2.
Collapse
Affiliation(s)
- Yafei Qu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weili Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Maggie Z X Xiao
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Yuejuan Zheng
- The Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Disease and Biosecurity, Shanghai University of Traditional Medicine, Shanghai, China
- Center for Traditional Chinese Medicine and Immunology Research, School of Basic Medical Sciences, Shanghai University of Traditional Medicine, Shanghai, China
| | - Qiming Liang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
42
|
Hammoudeh N, Soukkarieh C, Murphy DJ, Hanano A. Mammalian lipid droplets: structural, pathological, immunological and anti-toxicological roles. Prog Lipid Res 2023; 91:101233. [PMID: 37156444 DOI: 10.1016/j.plipres.2023.101233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Mammalian lipid droplets (LDs) are specialized cytosolic organelles consisting of a neutral lipid core surrounded by a membrane made up of a phospholipid monolayer and a specific population of proteins that varies according to the location and function of each LD. Over the past decade, there have been significant advances in the understanding of LD biogenesis and functions. LDs are now recognized as dynamic organelles that participate in many aspects of cellular homeostasis plus other vital functions. LD biogenesis is a complex, highly-regulated process with assembly occurring on the endoplasmic reticulum although aspects of the underpinning molecular mechanisms remain elusive. For example, it is unclear how many enzymes participate in the biosynthesis of the neutral lipid components of LDs and how this process is coordinated in response to different metabolic cues to promote or suppress LD formation and turnover. In addition to enzymes involved in the biosynthesis of neutral lipids, various scaffolding proteins play roles in coordinating LD formation. Despite their lack of ultrastructural diversity, LDs in different mammalian cell types are involved in a wide range of biological functions. These include roles in membrane homeostasis, regulation of hypoxia, neoplastic inflammatory responses, cellular oxidative status, lipid peroxidation, and protection against potentially toxic intracellular fatty acids and lipophilic xenobiotics. Herein, the roles of mammalian LDs and their associated proteins are reviewed with a particular focus on their roles in pathological, immunological and anti-toxicological processes.
Collapse
Affiliation(s)
- Nour Hammoudeh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Denis J Murphy
- School of Applied Sciences, University of South Wales, Pontypridd, CF37 1DL, Wales, United Kingdom..
| | - Abdulsamie Hanano
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria (AECS), P.O. Box 6091, Damascus, Syria..
| |
Collapse
|
43
|
Bresgen N, Kovacs M, Lahnsteiner A, Felder TK, Rinnerthaler M. The Janus-Faced Role of Lipid Droplets in Aging: Insights from the Cellular Perspective. Biomolecules 2023; 13:912. [PMID: 37371492 PMCID: PMC10301655 DOI: 10.3390/biom13060912] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
It is widely accepted that nine hallmarks-including mitochondrial dysfunction, epigenetic alterations, and loss of proteostasis-exist that describe the cellular aging process. Adding to this, a well-described cell organelle in the metabolic context, namely, lipid droplets, also accumulates with increasing age, which can be regarded as a further aging-associated process. Independently of their essential role as fat stores, lipid droplets are also able to control cell integrity by mitigating lipotoxic and proteotoxic insults. As we will show in this review, numerous longevity interventions (such as mTOR inhibition) also lead to strong accumulation of lipid droplets in Saccharomyces cerevisiae, Caenorhabditis elegans, Drosophila melanogaster, and mammalian cells, just to name a few examples. In mammals, due to the variety of different cell types and tissues, the role of lipid droplets during the aging process is much more complex. Using selected diseases associated with aging, such as Alzheimer's disease, Parkinson's disease, type II diabetes, and cardiovascular disease, we show that lipid droplets are "Janus"-faced. In an early phase of the disease, lipid droplets mitigate the toxicity of lipid peroxidation and protein aggregates, but in a later phase of the disease, a strong accumulation of lipid droplets can cause problems for cells and tissues.
Collapse
Affiliation(s)
- Nikolaus Bresgen
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Melanie Kovacs
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Angelika Lahnsteiner
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Thomas Klaus Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mark Rinnerthaler
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| |
Collapse
|
44
|
Li X, Kang K, Shen L, Shen L, Zhou Y. Integrative Analysis of the Predictive Value of Perilipin Family on Clinical Significance, Prognosis and Immunotherapy of Glioma. Biomedicines 2023; 11:biomedicines11041009. [PMID: 37189627 DOI: 10.3390/biomedicines11041009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Gliomas are common tumors of the central nervous system. The PLINs family is widely involved in the regulation of lipid metabolism and has been associated with the development and invasive metastasis of various malignancies. However, the biological role of the PLINs family in gliomas is still unclear. TIMER and UALCAN were used to assess PLINs mRNA expression in gliomas. “Survminer” and “Survival” were used to evaluate the connection between PLINs expression and glioma patients’ survival. cBioPortal was applied to assess PLINs’ genetic alterations in glioblastoma multiforme (GBM) and low-grade glioma (LGG). The correlation of PLINs expression with tumor immune cells was analyzed by TIMER. The expressions of PLIN1, PLIN4, and PLIN5 were decreased in GBM compared to normal tissues. However, PLIN2 and PLIN3 were significantly increased in GBM. Prognostic analysis showed that LGG patients with high PLIN1 expression had better overall survival (OS), and high expression of PLIN2/3/4/5 was associated with unfavorable OS. We further determined that the expression of PLINs members in gliomas was strongly related to tumor immune cells and immune checkpoint-associated genes. PLINS may be potential biomarkers for regulating the tumor microenvironment and predicting the efficacy of immunotherapy. In addition, we determined that PLIN1 may affect glioma patients’ therapeutic sensitivity to temozolomide. Our results demonstrated the biological significance and clinical values of PLINs in gliomas and provide a basis for future in-depth exploration of the specific mechanisms of each member of PLINs in gliomas.
Collapse
|
45
|
Park CY, Kim D, Seo MK, Kim J, Choe H, Kim JH, Hong JP, Lee YJ, Heo Y, Kim HJ, Park HS, Jang YJ. Dysregulation of Lipid Droplet Protein Expression in Adipose Tissues and Association with Metabolic Risk Factors in Adult Females with Obesity and Type 2 Diabetes. J Nutr 2023; 153:691-702. [PMID: 36931749 DOI: 10.1016/j.tjnut.2023.01.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Adipocyte dysregulation of lipid droplet (LD) metabolism caused by altered expression of LD proteins contributes to obesity-related metabolic diseases. OBJECTIVES We aimed to investigate whether expression levels of PLIN1, CIDEA, and CIDEC were altered in adipose tissues of women with obesity and type 2 diabetes and whether their alterations were associated with metabolic risk factors. METHODS Normal-weight (NW; 18.5 kg/m2 < BMI ≤ 25 kg/m2; n = 43), nondiabetic obese (OB; BMI > 30 kg/m2; n = 38), and diabetic obese (OB/DM; BMI > 30 kg/m2, fasting glucose ≥ 126 mg/dL, HbA1c ≥ 6.5%; n = 22) women were recruited. Metabolic parameters were measured, and expressions of PLIN1, CIDEA, CIDEC, and obesity-related genes were quantified in abdominal subcutaneous (SAT) and visceral adipose tissues (VAT). Effects of proinflammatory cytokines, endoplasmic reticulum (ER) stress inducers, and metabolic improvement agents on LD protein gene expressions were investigated in human adipocytes. RESULTS PLIN1, CIDEA, and CIDEC expressions were lower in SAT and higher in VAT in OB subjects relative to NW subjects; however, they were suppressed in both fat depots in OB/DM subjects relative to OB (P < 0.05). Across the entire cohort, whereas VAT PLIN1 (r = 0.349) and CIDEC expressions (r = 0.282) were positively associated with BMI (P < 0.05), SAT PLIN1 (r = -0.390) and CIDEA expressions (r = -0.565) were inversely associated. After adjustment for BMI, some or all of the adipose LD protein gene expressions were negatively associated with fasting glucose (r = -0.259 or higher) and triglyceride levels (r = -0.284 or higher) and positively associated with UCP1 expression (r = 0.353 or higher) (P < 0.05). In adipocytes, LD protein gene expressions were 55-70% downregulated by increased proinflammatory cytokines and ER stress but 2-4-fold upregulated by the metabolic improvement agents exendin-4 and dapagliflozin (P < 0.05). CONCLUSIONS The findings suggest that reduction of adipose LD protein expression is involved in the pathogenesis of metabolic disorders in women with obesity and type 2 diabetes and that increasing LD protein expression in adipocytes could control development of metabolic disorders.
Collapse
Affiliation(s)
- Chan Yoon Park
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Department of Food & Nutrition, College of Health Science, The University of Suwon, Hwaseong-si, Gyeonggi-do, Republic of Korea
| | - Donguk Kim
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min Kyeong Seo
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jimin Kim
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea; Brexogen Research Center, Brexogen Inc., Seoul, Republic of Korea
| | - Han Choe
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jong-Hyeok Kim
- Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joon Pio Hong
- Department of Obstetrics and Gynecology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yeon Ji Lee
- Department of Family Medicine, Inha University School of Medicine, Incheon, Republic of Korea
| | - Yoonseok Heo
- Department of General Surgery, Inha University School of Medicine, Incheon, Republic of Korea
| | - Hwa Jung Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, Seoul, Republic of Korea
| | - Hye Soon Park
- Department of Family Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Yeon Jin Jang
- Department of Physiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
46
|
Awadh AA. The Role of Cytosolic Lipid Droplets in Hepatitis C Virus Replication, Assembly, and Release. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5156601. [PMID: 37090186 PMCID: PMC10121354 DOI: 10.1155/2023/5156601] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 04/25/2023]
Abstract
The hepatitis C virus (HCV) causes chronic hepatitis by establishing a persistent infection. Patients with chronic hepatitis frequently develop hepatic cirrhosis, which can lead to liver cancer-the progressive liver damage results from the host's immune response to the unresolved infection. The HCV replication process, including the entry, replication, assembly, and release stages, while the virus circulates in the bloodstream, it is intricately linked to the host's lipid metabolism, including the dynamic of the cytosolic lipid droplets (cLDs). This review article depicts how this interaction regulates viral cell tropism and aids immune evasion by coining viral particle characteristics. cLDs are intracellular organelles that store most of the cytoplasmic components of neutral lipids and are assumed to play an increasingly important role in the pathophysiology of lipid metabolism and host-virus interactions. cLDs are involved in the replication of several clinically significant viruses, where viruses alter the lipidomic profiles of host cells to improve viral life cycles. cLDs are involved in almost every phase of the HCV life cycle. Indeed, pharmacological modulators of cholesterol synthesis and intracellular trafficking, lipoprotein maturation, and lipid signaling molecules inhibit the assembly of HCV virions. Likewise, small-molecule inhibitors of cLD-regulating proteins inhibit HCV replication. Thus, addressing the molecular architecture of HCV replication will aid in elucidating its pathogenesis and devising preventive interventions that impede persistent infection and prevent disease progression. This is possible via repurposing the available therapeutic agents that alter cLDs metabolism. This review highlights the role of cLD in HCV replication.
Collapse
Affiliation(s)
- Abdullah A. Awadh
- Department of Basic Medical Sciences, College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah 21423, Saudi Arabia
- King Abdullah International Medical Research Center, Jeddah 21423, Saudi Arabia
| |
Collapse
|
47
|
Ofori EK, Letsu BS, Amponsah SK, Ahenkorah J, Crabbe S, Kwao-Zigah G, Oppong SY, Diaba-Nuhoho P, Amanquah SD. Impact of blood perilipin A levels on obesity and metabolic health. BMC Res Notes 2022; 15:367. [PMID: 36503541 PMCID: PMC9743615 DOI: 10.1186/s13104-022-06261-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Perilipin A is a common protein that coats lipid surfaces preventing them from being exposed to oxidative damage. Researchers have found little consistency in the relationship between perilipin A levels in the blood and body fat. This study was a cross-sectional observational that looked at circulating perilipin A levels and how they relate to metabolic health. RESULTS The participants in this study were 86 individuals with a mean age of 45.5 ± 1.2 years. Multiple clinical and metabolic indicators (age, weight, BMI, total body fat mass, triglyceride, and HOMA-IR) were shown to be inversely associated with perilipin A levels (rho = - 0.32, - 0.37, - 0.40, - 0.45, - 0.33 and - 0.29; p < 0.05 respectively). Obese persons were almost six times more likely than non-obese individuals to have lower perilipin A levels (odds ratio = 6.22, CI = 2.35-11.50, p < 0.001). Our findings underscore the important role of perilipin A proteins in metabolic health.
Collapse
Affiliation(s)
- Emmanuel K Ofori
- Department of Chemical Pathology, University of Ghana Medical School, Accra, Ghana.
| | - Bright Selorm Letsu
- Department of Chemical Pathology, University of Ghana Medical School, Accra, Ghana
| | - Seth K Amponsah
- Department of Medical Pharmacology, University of Ghana Medical School, Accra, Ghana
| | - John Ahenkorah
- Department of Anatomy, University of Ghana Medical School, Accra, Ghana
| | | | - Genevieve Kwao-Zigah
- Department of Chemical Pathology, University of Ghana Medical School, Accra, Ghana
| | | | | | - Seth D Amanquah
- Department of Chemical Pathology, University of Ghana Medical School, Accra, Ghana
| |
Collapse
|
48
|
Zhang E. Hepatic PLIN5 Deficiency Impairs Lipogenesis through Mitochondrial Dysfunction. Int J Mol Sci 2022; 23:15598. [PMID: 36555245 PMCID: PMC9779494 DOI: 10.3390/ijms232415598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/01/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Regulation of lipid droplets (LDs) metabolism is the core of controlling intracellular fatty acids (FAs) fluxes, and perilipin 5 (PLIN5) plays a key role in this process. Our previous studies have found that hepatic PLIN5 deficiency reduces LDs accumulation, but the trafficking of FAs produced from this pathway and the interaction between mitochondria and LDs in this process are largely unknown. Here, we found that the deficiency of PLIN5 decreases LDs accumulation by increasing FAs efflux. In addition, the decreased lipogenesis of PLIN5-deficient hepatocytes is accompanied by mitochondrial dysfunction, suggesting that PLIN5 plays an important role in mediating the interaction between LDs and mitochondria. Importantly, PLIN5 ablation negates oxidative capacity differences of peri-droplet and cytosolic mitochondria. In summary, these data indicate that PLIN5 plays a vital role in maintaining mitochondrial-mediated lipogenesis, which provides an important new perspective on the regulation of liver lipid storage and the relationship between PLIN5 and mitochondria.
Collapse
Affiliation(s)
- Enxiang Zhang
- Key Laboratory of Growth Regulation and Transformation Research of Zhejiang Province, School of Life Sciences, Westlake Institute for Advanced Study, Westlake University, Hangzhou 310024, China;
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, USA
- Duke Regeneration Center, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
49
|
Wu Y, Chen K, Li L, Hao Z, Wang T, Liu Y, Xing G, Liu Z, Li H, Yuan H, Lu J, Zhang C, Zhang J, Zhao D, Wang J, Nie J, Ye D, Pan G, Chan WY, Liu X. Plin2-mediated lipid droplet mobilization accelerates exit from pluripotency by lipidomic remodeling and histone acetylation. Cell Death Differ 2022; 29:2316-2331. [PMID: 35614132 PMCID: PMC9613632 DOI: 10.1038/s41418-022-01018-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/07/2022] [Accepted: 05/08/2022] [Indexed: 12/29/2022] Open
Abstract
Metabolic switch is critical for cell fate determination through metabolic functions, epigenetic modifications, and gene expression. However, the mechanisms underlying these alterations and their functional roles remain unclear. Here, we show that Plin2-mediated moderate lipid hydrolysis is critical for pluripotency of embryonic stem cells (ESCs). Upon exit from pluripotency, lipid droplet (LD)-associated protein Plin2 is recognized by Hsc70 and degraded via chaperone-mediated autophagy to facilitate LD mobilization. Enhancing lipid hydrolysis by Plin2 knockout promotes pluripotency exit, which is recovered by ATGL inhibition. Mechanistically, excessive lipid hydrolysis induces a dramatic lipidomic remodeling characterized by decreased cardiolipin and phosphatidylethanolamine, which triggers defects in mitochondrial cristae and fatty acid oxidation, resulting in reduced acetyl-CoA and histone acetylation. Our results reveal how LD mobilization is regulated and its critical role in ESC pluripotency, and indicate the mechanism linking LD homeostasis to mitochondrial remodeling and epigenetic regulation, which might shed light on development and diseases.
Collapse
Affiliation(s)
- Yi Wu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Keshi Chen
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Linpeng Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zhihong Hao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tianyu Wang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Yang Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guangsuo Xing
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Zichao Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Heying Li
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Hao Yuan
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jianghuan Lu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | | | | | - Danyun Zhao
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Junwei Wang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jinfu Nie
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Dan Ye
- Fudan University, Shanghai, 200433, China
| | - Guangjin Pan
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Wai-Yee Chan
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, China-New Zealand Joint Laboratory on Biomedicine and Health, CUHK-GIBH Joint Research Laboratory on Stem Cells and Regenerative Medicine, Institute for Stem Cell and Regeneration, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Centre for Regenerative Medicine and Health, Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong SAR, China.
| |
Collapse
|
50
|
Al Saedi A, Debruin DA, Hayes A, Hamrick M. Lipid metabolism in sarcopenia. Bone 2022; 164:116539. [PMID: 36007811 DOI: 10.1016/j.bone.2022.116539] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/10/2022] [Accepted: 08/18/2022] [Indexed: 11/29/2022]
Abstract
Sarcopenia is an age-related disease associated with loss of muscle mass and strength. This geriatric syndrome predisposes elderly individuals to a disability, falls, fractures, and death. Fat infiltration in muscle is one of the hallmarks of sarcopenia and aging. Alterations in fatty acid (FA) metabolism are evident in aging, type 2 diabetes, and obesity, with the accumulation of lipids inside muscle cells contributing to muscle insulin resistance and ceramide accumulation. These lipids include diacylglycerol, lipid droplets, intramyocellular lipids, intramuscular triglycerides, and polyunsaturated fatty acids (PUFAs). In this review, we examine the regulation of lipid metabolism in skeletal muscle, including lipid metabolization and storage, intervention, and the types of lipases expressed in skeletal muscle responsible for the breakdown of adipose triglyceride fats. In addition, we address the role of FAs in sarcopenia and the potential benefits of PUFAs.
Collapse
Affiliation(s)
- Ahmed Al Saedi
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia; Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, VIC, Australia; Institute of Health and Sport (IHeS), Victoria University, Melbourne, VIC, Australia.
| | - Danielle A Debruin
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia; Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, VIC, Australia; Institute of Health and Sport (IHeS), Victoria University, Melbourne, VIC, Australia
| | - Alan Hayes
- Australian Institute for Musculoskeletal Science (AIMSS), The University of Melbourne and Western Health, St. Albans, VIC, Australia; Department of Medicine-Western Health, Melbourne Medical School, The University of Melbourne, St. Albans, VIC, Australia; Institute of Health and Sport (IHeS), Victoria University, Melbourne, VIC, Australia
| | - Mark Hamrick
- Department of Cellular Biology & Anatomy, Medical College of Georgia, Augusta University, Laney Walker Blvd. CB2915, Augusta, GA 30912, USA
| |
Collapse
|