1
|
Tian X, Ran Z, Yan B, Zhu J, Zhou Q, Kong F, Yan X, Xu J. Lipid droplets play versatile roles in ovarian development of the razor clam Sinonovacula constricta: Insights from proteomic and lipidomic analyses. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2025; 55:101519. [PMID: 40288072 DOI: 10.1016/j.cbd.2025.101519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 04/21/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Lipid droplet (LD) deposition is a common phenomenon during ovarian development across marine mollusks; however, studies on the protein and lipid composition of their ovarian LDs remain limited. Here, we purified LDs from the ovaries of Sinonovacula constricta and isolated proteins and lipids from these purified LDs for proteomic and lipidomic analyses. Our proteomic analysis identified 3243 proteins, with PLIN2 being the most abundant (37.03 ± 13.56 %). We subsequently conducted a functional analysis of the top 500 most abundant LD-associated proteins, categorizing them into 15 groups, including those involved in lipid metabolism, sterol biosynthesis, tricarboxylic acid cycle, carbohydrate metabolism, G protein superfamily, protein chaperones, transport proteins, nucleotide-catabolic process, protein processing and degradation, cytoskeletal proteins, oxidative stress and immunity, and ribosome-associated proteins. In our lipidomic analysis, we identified 1158 molecules across 52 lipid classes, with phosphatidylcholine (PC) exhibiting the greatest diversity at 209 varieties, followed by EtherPC with 177 varieties and triglyceride (TG) with 149 varieties. The fatty acid (FA) analysis of LDs revealed that 16:0 was the most abundant (30.01 ± 0.42 %). Additionally, LDs were found rich in long-chain polyunsaturated FAs (35.63 ± 4.36 %), particularly EPA and DHA. Moreover, we analyzed the FA composition of TGs, PCs, and EtherPCs derived from ovarian LDs. In PCs and TGs, the predominant FAs were 16:0, 16:1, and 18:3, while 16:0, 22:6, and 18:4 constituted the major FA species in EtherPCs. Together, our results suggest that ovarian LDs in S. constricta not only participate in lipid metabolism but also interact with other organelles and metabolic processes, thereby facilitating ovarian development.
Collapse
Affiliation(s)
- Xuxu Tian
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Zhaoshou Ran
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo 315211, China; Ningbo Yongyuan Aquatic Products Co Ltd, Ningbo 315601, China.
| | - Bowen Yan
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Jiaxin Zhu
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Qiang Zhou
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Fei Kong
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Xiaojun Yan
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Jilin Xu
- Key Laboratory of Aquacultural Biotechnology, Ministry of Education, Ningbo University, Ningbo 315211, China; Key Laboratory of Marine Biotechnology of Zhejiang Province, Ningbo 315211, China; Ningbo Yongyuan Aquatic Products Co Ltd, Ningbo 315601, China; Fujian Dalai Seeding Technology Co Ltd, Fuzhou 350600, China.
| |
Collapse
|
2
|
Zhu L, Ming H, Scatolin GN, Xiao A, Jiang Z. METTL7A improves bovine IVF embryo competence by attenuating oxidative stress†. Biol Reprod 2025; 112:628-639. [PMID: 39883095 PMCID: PMC11996759 DOI: 10.1093/biolre/ioaf018] [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: 08/16/2024] [Revised: 12/17/2024] [Accepted: 01/28/2025] [Indexed: 01/31/2025] Open
Abstract
In vitro fertilization is a widely used assisted reproductive technology to achieve a successful pregnancy. However, the acquisition of oxidative stress in embryo in vitro culture impairs its competence. Here, we demonstrated that a nuclear coding gene, methyltransferase-like protein 7A, improves the developmental potential of bovine embryos. We found that exogenous methyltransferase-like protein 7A modulates expression of genes involved in embryonic cell mitochondrial pathways and promotes trophectoderm development. Surprisingly, we discovered that methyltransferase-like protein 7A alleviates mitochondrial stress and DNA damage and promotes cell cycle progression during embryo cleavage. In summary, we have identified a novel mitochondria stress eliminating mechanism regulated by methyltransferase-like protein 7A that occurs during the acquisition of oxidative stress in embryo in vitro culture. This discovery lays the groundwork for the development of methyltransferase-like protein 7A as a promising therapeutic target for in vitro fertilization embryo competence.
Collapse
Affiliation(s)
- Linkai Zhu
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Hao Ming
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Giovanna N Scatolin
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Andrew Xiao
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - Zongliang Jiang
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
3
|
Cui L, Mai Z, Lu Y, Zheng J, Lin P, Chen X, Zheng Y, Lin Y, Guo B, Zhao X. Laboratory investigation of METTL7A driving MSC osteogenic differentiation through YAP1 translation enhancement via eIF4F recruitment. Int Endod J 2025; 58:587-603. [PMID: 39815670 DOI: 10.1111/iej.14198] [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: 09/06/2024] [Revised: 01/07/2025] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
AIM Effective control of mesenchymal stem cell (MSC) differentiation towards osteogenic lineages is fundamental for bone regeneration. This study elucidates the regulatory role of methyltransferase like 7A (METTL7A) in the osteogenic differentiation of MSCs. METHODOLOGY Alkaline phosphatase staining, Alizarin Red S staining, western blotting, and in vivo studies were conducted to determine the effects of METTL7A depletion or overexpression on the osteogenic differentiation of various types of MSCs. Then the downstream signalling pathways regulated by METTL7A in MSCs were further investigated. RESULTS Our findings indicate that METTL7A expression significantly increases during the osteogenic differentiation of MSCs. Furthermore, depletion of METTL7A hindered, whereas its overexpression enhanced, the osteogenic differentiation of MSCs. Mechanistically, METTL7A influences MSC osteogenic differentiation by activating the YAP1-TEAD1 signalling pathway. It enhances YAP1 expression not only by stabilising YAP1 mRNA but also, crucially, by recruiting the eIF4F complex, thereby boosting the translation efficiency of YAP1 mRNA. Additionally, the YAP1/TEAD1 complex transcriptionally regulates METTL7A expression, creating a positive feedback loop that amplifies osteogenic differentiation. CONCLUSIONS Overall, our study uncovers a previously unknown molecular mechanism of MSC osteogenic differentiation and suggests that activating METTL7A could offer new avenues for enhancing bone regeneration.
Collapse
Affiliation(s)
- Li Cui
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, California, USA
| | - Zizhao Mai
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ye Lu
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Pei Lin
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xu Chen
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yucheng Zheng
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunfan Lin
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bing Guo
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xinyuan Zhao
- School of Stomatology, Stomatological Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
4
|
Kitamoto T, Kitamoto A. Integrative proteomic and lipidomic analysis of GNB1 and SCARB2 knockdown in human subcutaneous adipocytes. PLoS One 2025; 20:e0319163. [PMID: 40127054 PMCID: PMC11932494 DOI: 10.1371/journal.pone.0319163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/28/2025] [Indexed: 03/26/2025] Open
Abstract
Obesity, a global public health concern, is influenced by various factors, including genetic predispositions. Although many obesity-associated genes have been identified through genome-wide association studies (GWAS), the molecular mechanisms linking these genes to adipose tissue function remain largely unexplored. This study integrates proteomic data on adipocyte fat accumulation with GWAS data on obesity to unravel the roles of the identified key candidate genes - G protein subunit beta 1 (GNB1) and scavenger receptor class B member 2 (SCARB2) - involved in fat accumulation. We utilized RNA interference to knock down GNB1 and SCARB2 in human subcutaneous adipocytes, followed by lipidome and proteome analyses using mass spectrometry. Knockdown of these genes resulted in a reduction in lipid droplet accumulation, indicating their role in adipocyte lipid storage. Digital PCR confirmed effective gene knockdown, with GNB1 and SCARB2 mRNA levels significantly reduced. In total, the lipidomic analysis identified 96 lipid species with significant alterations. GNB1 knockdown resulted in a decrease in cholesterol esters and an increase in phosphatidylcholines, phosphatidylinositols, and ceramides. SCARB2 knockdown also led to an increase in phosphatidylcholines, with a trend towards decreased triacylglycerols. Proteomic analysis revealed significant changes in proteins involved in lipid metabolism and adipocyte function, including PLPP1 and CDH13, which were upregulated following GNB1 knockdown, and HSPA8, which was downregulated. Conversely, SCARB2 knockdown resulted in the downregulation of PLPP1 and METTL7A, and the upregulation of PLIN2, HSPA8, NPC2, and SQSTM1. Our findings highlight the significant roles of GNB1 and SCARB2 in lipid metabolism and adipocyte function, providing insights that could inform therapeutic strategies targeting these regulatory genes in obesity.
Collapse
Affiliation(s)
- Takuya Kitamoto
- Advanced Research Facilities and Services, Division of Preeminent Research Supports, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Aya Kitamoto
- Advanced Research Facilities and Services, Division of Preeminent Research Supports, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
5
|
Starble RM, Sun EG, Gbyli R, Radda J, Lu J, Jensen TB, Sun N, Khudaverdyan N, Hu B, Melnick MA, Zhao S, Roper N, Wang GG, Song J, Politi K, Wang S, Xiao AZ. Epigenetic priming promotes acquisition of tyrosine kinase inhibitor resistance and oncogene amplification in human lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.26.634826. [PMID: 39974875 PMCID: PMC11838195 DOI: 10.1101/2025.01.26.634826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
In mammalian cells, gene copy number is tightly controlled to maintain gene expression and genome stability. However, a common molecular feature across cancer types is oncogene amplification, which promotes cancer progression by drastically increasing the copy number and expression of tumor-promoting genes. For example, in tyrosine kinase inhibitor (TKI)-resistant lung adenocarcinoma (LUAD), oncogene amplification occurs in over 40% of patients' tumors. Despite the prevalence of oncogene amplification in TKI-resistant tumors, the mechanisms facilitating oncogene amplification are not fully understood. Here, we find that LUADs exhibit a unique chromatin signature demarcated by strong CTCF and cohesin deposition in drug-naïve tumors, which correlates with the boundaries of oncogene amplicons in TKI-resistant LUAD cells. We identified a global chromatin priming effect during the acquisition of TKI resistance, marked by a dynamic increase of H3K27Ac, cohesin loading, and inter-TAD interactions, which occurs before the onset of oncogene amplification. Furthermore, we have found that the METTL7A protein, which was previously reported to localize to the endoplasmic reticulum and inner nuclear membrane, has a novel chromatin regulatory function by binding to amplified loci and regulating cohesin recruitment and inter-TAD interactions. Surprisingly, we discovered that METTL7A remodels the chromatin landscape prior to large-scale copy number gains. Furthermore, while METTL7A depletion has little effect on the chromatin structure and proliferation of drug-naïve cells, METTL7A depletion prevents the formation and maintenance of TKI resistant-clones, highlighting the specific role of METTL7A as cells are becoming resistant. In summary, we discovered an unexpected mechanism required for the acquisition of TKI resistance regulated by a largely uncharacterized factor, METTL7A. This discovery sheds light into the maintenance of oncogene copy number and paves the way to the development of new therapeutics for preventing TKI resistance in LUAD.
Collapse
Affiliation(s)
- Rebecca M Starble
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Eric G Sun
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Present address: Tri-Institutional MD-PhD Program, Weill Cornell Medicine, Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Rana Gbyli
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jonathan Radda
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
| | - Jiuwei Lu
- Department of Biochemistry, University of California, Riverside, CA 92521, USA
| | - Tyler B Jensen
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| | - Ning Sun
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
- Present address: Key Laboratory of Growth Regulation and Transformation Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Nelli Khudaverdyan
- Department of Biochemistry, University of California, Riverside, CA 92521, USA
| | - Bomiao Hu
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | | | - Shuai Zhao
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Nitin Roper
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Gang Greg Wang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jikui Song
- Department of Biochemistry, University of California, Riverside, CA 92521, USA
| | - Katerina Politi
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Cancer Center, New Haven, CT 06520, USA
- Department of Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT 06510, USA
| | - Siyuan Wang
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Andrew Z Xiao
- Department of Genetics, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
6
|
Zhu L, Ming H, Scatolin GN, Xiao A, Jiang Z. METTL7A improves bovine IVF embryo competence by attenuating oxidative stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.17.628915. [PMID: 39763908 PMCID: PMC11702634 DOI: 10.1101/2024.12.17.628915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
In vitro fertilization (IVF) is a widely used assisted reproductive technology to achieve a successful pregnancy. However, the acquisition of oxidative stress in embryo in vitro culture impairs its competence. Here, we demonstrated that a nuclear coding gene, methyltransferase-like protein 7A (METTL7A), improves the developmental potential of bovine embryos. We found that exogenous METTL7A modulates expression of genes involved in embryonic cell mitochondrial pathways and promotes trophectoderm development. Surprisingly, we discovered that METTL7A alleviates mitochondrial stress and DNA damage and promotes cell cycle progression during embryo cleavage. In summary, we have identified a novel mitochondria stress eliminating mechanism regulated by METTL7A that occurs during the acquisition of oxidative stress in embryo in vitro culture. This discovery lays the groundwork for the development of METTL7A as a promising therapeutic target for IVF embryo competence.
Collapse
Affiliation(s)
- Linkai Zhu
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Hao Ming
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Giovanna N. Scatolin
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| | - Andrew Xiao
- Department of Genetics, School of Medicine, Yale University, New Haven, CT, USA
| | - Zongliang Jiang
- Department of Animal Sciences, Genetics Institute, University of Florida, Gainesville, FL, USA
| |
Collapse
|
7
|
Lian C, Li F, Xie Y, Zhang L, Chen H, Wang Z, Pan X, Wang X, Zhang J. Identification of T-cell exhaustion-related genes and prediction of their immunotherapeutic role in lung adenocarcinoma. J Cancer 2024; 15:2160-2178. [PMID: 38495503 PMCID: PMC10937285 DOI: 10.7150/jca.92839] [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: 12/02/2023] [Accepted: 02/06/2024] [Indexed: 03/19/2024] Open
Abstract
Background: Lung adenocarcinoma ranks as the second most widespread form of cancer globally, accompanied by a significant mortality rate. Several studies have shown that T cell exhaustion is associated with immunotherapy of tumours. Consequently, it is essential to comprehend the possible impact of T cell exhaustion on the tumor microenvironment. The purpose of this research was to create a TEX-based model that would use single-cell RNA-seq (scRNA-seq) and bulk-RNA sequencing to explore new possibilities for assessing the prognosis and immunotherapeutic response of LUAD patients. Methods: RNA-seq data from LUAD patients was downloaded from the Cancer Genome Atlas (TCGA) database and the National Center for Biotechnology Information (GEO). 10X scRNA sequencing data, as reported by Bischoff P et al., was utilized for down-sampling clustering and subgroup identification using TSNE. TEX-associated genes were identified through gene set variance analysis (GSVA) and weighted gene correlation network analysis (WGCNA). We utilized LASSO-Cox analysis to establish predicted TEX features. External validation was conducted in GSE31210 and GSE30219 cohorts. Immunotherapeutic response was assessed in IMvigor210, GSE78220, GSE35640 and GSE100797 cohorts. Furthermore, we investigated differences in mutational profiles and immune microenvironment between various risk groups. We then screened TEXRS key regulatory genes using ROC diagnostic curves and KM survival curves. Finally, we verified the differential expression of key regulatory genes through RT-qPCR. Results: Nine TEX genes were identified as highly predictive of LUAD prognosis and strongly correlated with disease outcome. Univariate and multivariate analysis revealed that patients in the low-risk group had significantly better overall survival rates compared with those in the high-risk group, highlighting the model's ability to independently predict LUAD prognosis. Our analysis revealed significant variation in the biological function, mutational landscape, and immune cell infiltration within the tumor microenvironment of both high-risk and low-risk groups. Additionally, immunotherapy was found to have a significant impact on both groups, indicating strong predictive efficacy of the model. Conclusions: The TEX model showed good predictive performance and provided a new perspective for evaluating the efficacy of preimmunization, which provides a new strategy for the future treatment of lung adenocarcinoma.
Collapse
Affiliation(s)
- Chaoqun Lian
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu 233030, China
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Feifan Li
- Department of Tumor Radiotherapy, The First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Yiluo Xie
- Department of Clinical Medicine, Bengbu Medical University, Bengbu 233030, China
| | - Linxiang Zhang
- Research Center of Clinical Laboratory Science, Bengbu Medical University, Bengbu 233030, China
| | - Huili Chen
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Ziqiang Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Xinyu Pan
- Department of Medical Imaging, Bengbu Medical University, Bengbu 233030, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Molecular Diagnosis Center, Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu 233030, China
| | - Jing Zhang
- Department of Genetics, School of Life Sciences, Bengbu Medical University, Bengbu 233030, China
| |
Collapse
|
8
|
Wang B, Jiang J, Luo D, Wang X. Pan-cancer analysis reveals potential immunological and prognostic roles of METTL7A in human cancers. Sci Rep 2024; 14:3476. [PMID: 38342956 PMCID: PMC10859372 DOI: 10.1038/s41598-024-54255-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 02/10/2024] [Indexed: 02/13/2024] Open
Abstract
Methyltransferase-like protein 7A (METTL7A) is an m6A RNA methyltransferase that has been linked to cancer prognosis and drug resistance. However, a comprehensive analysis of METTL7A is lacking. The expression of METTL7A, prognostic performance, correlation with microsatellite instability (MSI), tumor mutational burden (TMB), and immune infiltration was investigated in The Cancer Genome Atlas (TCGA). Immunohistochemistry staining was applied to detect METTL7A in 6 tumors. METTL7A was significantly decreased in 19 cancers in TCGA including LUAD. Alterations of METTL7A include amplification and mutation, and epigenetic alterations revealed increased promoter methylation may result in down-regulation of METTL7A in LUAD. We also found that METTL7A was linked to both TMB and MSI in LUAD. METTL7A was increasingly correlated with invasive immune cells, while being negatively associated with Macrophages M0, Mast cells activated, activated memory CD4 T cells, CD8 T cells, and follicular helper T cells in several tumors. Additionally, METTL7A showed similar correlation with immune therapy-related genes across cancers. Our biological validation found that the protein levels of METTL7A were down-regulated in breast cancer (BRCA), endometrioid cancer (UCEC), colon cancer (COAD), prostate cancer (PRAD), and kidney clear cell carcinoma (KIRC), as detected by immunohistochemistry staining. Overall, our work indicates that METTL7A may serve as promising diagnostic and prognostic indicator of LUAD, and our work sheds light on the potential immunological and prognostic roles of METTL7A in human cancers.
Collapse
Affiliation(s)
- Bin Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Jiang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danju Luo
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiong Wang
- Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
9
|
Khaddaj R, Stribny J, Cottier S, Schneiter R. Perilipin 3 promotes the formation of membrane domains enriched in diacylglycerol and lipid droplet biogenesis proteins. Front Cell Dev Biol 2023; 11:1116491. [PMID: 37465010 PMCID: PMC10350540 DOI: 10.3389/fcell.2023.1116491] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/14/2023] [Indexed: 07/20/2023] Open
Abstract
Lipid droplets (LDs) serve as intracellular stores of energy-rich neutral lipids. LDs form at discrete sites in the endoplasmic reticulum (ER) and they remain closely associated with the ER during lipogenic growth and lipolytic consumption. Their hydrophobic neutral lipid core is covered by a monolayer of phospholipids, which harbors a specific set of proteins. This LD surface is coated and stabilized by perilipins, a family of soluble proteins that specifically target LDs from the cytosol. We have previously used chimeric fusion proteins between perilipins and integral ER membrane proteins to test whether proteins that are anchored to the ER bilayer could be dragged onto the LD monolayer. Expression of these chimeric proteins induces repositioning of the ER membrane around LDs. Here, we test the properties of membrane-anchored perilipins in cells that lack LDs. Unexpectedly, membrane-anchored perilipins induce expansion and vesiculation of the perinuclear membrane resulting in the formation of crescent-shaped membrane domains that harbor LD-like properties. These domains are stained by LD-specific lipophilic dyes, harbor LD marker proteins, and they transform into nascent LDs upon induction of neutral lipid synthesis. These ER domains are enriched in diacylglycerol (DAG) and in ER proteins that are important for early steps of LD biogenesis, including seipin and Pex30. Formation of the domains in vivo depends on DAG levels, and we show that perilipin 3 (PLIN3) binds to liposomes containing DAG in vitro. Taken together, these observations indicate that perilipin not only serve to stabilize the surface of mature LDs but that they are likely to exert a more active role in early steps of LD biogenesis at ER subdomains enriched in DAG, seipin, and neutral lipid biosynthetic enzymes.
Collapse
Affiliation(s)
- Rasha Khaddaj
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Jiri Stribny
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Stéphanie Cottier
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
10
|
Vera-Montecinos A, Galiano-Landeira J, Roldán M, Vidal-Domènech F, Claro E, Ramos B. A Novel Localization of METTL7A in Bergmann Glial Cells in Human Cerebellum. Int J Mol Sci 2023; 24:ijms24098405. [PMID: 37176112 PMCID: PMC10179429 DOI: 10.3390/ijms24098405] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 04/28/2023] [Accepted: 05/01/2023] [Indexed: 05/15/2023] Open
Abstract
Methyltransferase-like protein 7A (METTL7A) is a member of the METTL family of methyltransferases.Little information is available regarding the cellular expression of METTL7A in the brain. METTL7A is commonly located in the endoplasmic reticulum and to a lesser extent, in the lipid droplets of some cells. Several studies have reported altered protein and RNA levels in different brain areas in schizophrenia. One of these studies found reduced protein levels of METTL7A in the cerebellar cortex in schizophrenia and stress murine models. Since there is limited information in the literature about METTL7A, we characterized its cellular and subcellular localizations in the human cerebellum using immunohistochemical analysis with laser confocal microscopy. Our study reveals a novel METTL7A localization in GFAP-positive cells, with higher expression in the end-feet of the Bergmann glia, which participate in the cerebrospinal fluid-brain parenchyma barrier. Further 3D reconstruction image analysis showed that METTL7A was expressed in the contacts between the Bergmann glia and Purkinje neurons. METTL7A was also detected in lipid droplets in some cells in the white matter. The localization of METTL7A in the human cerebellar glia limitans could suggest a putative role in maintaining the cerebellar parenchyma homeostasis and in the regulation of internal cerebellar circuits by modulating the synaptic activity of Purkinje neurons.
Collapse
Affiliation(s)
- América Vera-Montecinos
- Psiquiatria Molecular, Parc Sanitari Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain
| | - Jordi Galiano-Landeira
- Psiquiatria Molecular, Parc Sanitari Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain
| | - Mònica Roldán
- Unitat de Microscòpia Confocal i Imatge Cel·lular, Servei de Medicina Genètica i Molecular, Institut Pediàtric de Malalties Rares (IPER), Hospital Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
- Institut de Recerca Sant Joan de Déu, Esplugues de Llobregat, 08950 Barcelona, Spain
| | - Francisco Vidal-Domènech
- Psiquiatria Molecular, Parc Sanitari Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain
| | - Enrique Claro
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Belén Ramos
- Psiquiatria Molecular, Parc Sanitari Sant Joan de Déu, Institut de Recerca Sant Joan de Déu, Dr. Antoni Pujadas, 42, 08830 Sant Boi de Llobregat, Spain
- Departament de Bioquímica i Biologia Molecular, Facultat de Medicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Centro de Investigación Biomédica en Red de Salud Mental, CIBERSAM (Biomedical Network Research Center of Mental Health), Institute of Health Carlos III, 28029 Madrid, Spain
- Faculty of Medicine, University of Vic-Central University of Catalonia, 08500 Vic, Spain
| |
Collapse
|
11
|
Evidence Based on an Integrative Analysis of Multi-Omics Data on METTL7A as a Molecular Marker in Pan-Cancer. Biomolecules 2023; 13:biom13020195. [PMID: 36830565 PMCID: PMC9952925 DOI: 10.3390/biom13020195] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Methyltransferase-like protein 7A (METTL7A), an RNA N6-methyladenosine (m6A) methyltransferase, has attracted much attention as it has been found to be closely associated with various types of tumorigenesis and progression. This study provides a comprehensive assessment of METTL7A from a pan-cancer perspective using multi-omics data. The gene ontology enrichment analysis of METTL7A-binding proteins revealed a close association with methylation and lipid metabolism. We then explored the expression of METTL7A in normal tissues, cell lines, different subtypes and cancers, and found that METTL7A was differentially expressed in various cancer species, tumor molecular subtypes and immune subtypes. Evaluation of the diagnostic and prognostic value of METTL7A in pan-cancer revealed that METTL7A had high accuracy in tumor prediction. Moreover, the low expression of METTL7A significantly correlated with the poor prognosis, including kidney renal clear cell carcinoma (KIRC), mesothelioma and sarcoma, indicating that METTL7A could be a potential biomarker for tumor diagnosis and prognosis. We focused on KIRC after pre-screening and analyzed its expression and prognostic value in various clinical subgroups. We found that METTL7A was significantly related to tumor stage, metastasis stage, pathologic stage, primary therapy outcome, histologic grade and gender, and that low METTL7A expression was associated with poorer outcomes. Finally, we analyzed the immune infiltration and co-expressed genes of METTL7A as well as the differentially expressed genes in the high and low expression groups. In conclusion, METTL7A is a better molecular marker for pan-cancer diagnosis and prognosis and has high potential as a diagnostic and prognostic biomarker for KIRC.
Collapse
|
12
|
Kochenova OV, Mukkavalli S, Raman M, Walter JC. Cooperative assembly of p97 complexes involved in replication termination. Nat Commun 2022; 13:6591. [PMID: 36329031 PMCID: PMC9633789 DOI: 10.1038/s41467-022-34210-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022] Open
Abstract
The p97 ATPase extracts polyubiquitylated proteins from diverse cellular structures in preparation for destruction by the proteasome. p97 functions with Ufd1-Npl4 and a variety of UBA-UBX co-factors, but how p97 complexes assemble on ubiquitylated substrates is unclear. To address this, we investigated how p97 disassembles the CMG helicase after it is ubiquitylated during replication termination. We show that p97Ufd1-Npl4 recruitment to CMG requires the UBA-UBX protein Ubxn7, and conversely, stable Ubxn7 binding to CMG requires p97Ufd1-Npl4. This cooperative assembly involves interactions between Ubxn7, p97, Ufd1-Npl4, and ubiquitin. Another p97 co-factor, Faf1, partially compensates for the loss of Ubxn7. Surprisingly, p97Ufd1-Npl4-Ubxn7 and p97Ufd1-Npl4-Faf1 also assemble cooperatively on unanchored ubiquitin chains. We propose that cooperative and substrate-independent recognition of ubiquitin chains allows p97 to recognize an unlimited number of polyubiquitylated proteins while avoiding the formation of partial, inactive complexes.
Collapse
Affiliation(s)
- Olga V Kochenova
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Sirisha Mukkavalli
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Malavika Raman
- Department of Developmental Molecular and Chemical Biology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Johannes C Walter
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Blavatnik Institute, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
13
|
Zheng J, Cao Y, Yang J, Jiang H. UBXD8 mediates mitochondria-associated degradation to restrain apoptosis and mitophagy. EMBO Rep 2022; 23:e54859. [PMID: 35979733 PMCID: PMC9535754 DOI: 10.15252/embr.202254859] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/20/2022] [Accepted: 08/04/2022] [Indexed: 08/08/2023] Open
Abstract
The hexameric AAA-ATPase valosin-containing protein (VCP) is essential for mitochondrial protein quality control. How VCP is recruited to mammalian mitochondria remains obscure. Here we report that UBXD8, an ER- and lipid droplet-localized VCP adaptor, also localizes to mitochondria and locally recruits VCP. UBXD8 associates with mitochondrial and ER ubiquitin E3 ligases and targets their substrates for degradation. Remarkably, both mitochondria- and ER-localized UBXD8 can degrade mitochondrial and ER substrates in cis and in trans. UBXD8 also associates with the TOM complex but is dispensable for translocation-associated degradation. UBXD8 knockout impairs the degradation of the pro-survival protein Mcl1 but surprisingly sensitizes cells to apoptosis and mitochondrial stresses. UBXD8 knockout also hyperactivates mitophagy. We identify pro-apoptotic BH3-only proteins Noxa, Bik, and Bnip3 as novel UBXD8 substrates and determine that UBXD8 inhibits apoptosis via degrading Noxa and restrains mitophagy via degrading Bnip3. Collectively, our characterizations reveal UBXD8 as the major mitochondrial adaptor of VCP and unveil its role in apoptosis and mitophagy regulation.
Collapse
Affiliation(s)
- Jing Zheng
- School of Life SciencesPeking UniversityBeijingChina
- National Institute of Biological SciencesBeijingChina
- Beijing Key Laboratory of Cell Biology for Animal AgingBeijingChina
| | - Yu Cao
- National Institute of Biological SciencesBeijingChina
- Beijing Key Laboratory of Cell Biology for Animal AgingBeijingChina
- College of Life SciencesBeijing Normal UniversityBeijingChina
| | - Jun Yang
- National Institute of Biological SciencesBeijingChina
- Beijing Key Laboratory of Cell Biology for Animal AgingBeijingChina
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Hui Jiang
- School of Life SciencesPeking UniversityBeijingChina
- National Institute of Biological SciencesBeijingChina
- Beijing Key Laboratory of Cell Biology for Animal AgingBeijingChina
- Tsinghua Institute of Multidisciplinary Biomedical ResearchTsinghua UniversityBeijingChina
| |
Collapse
|
14
|
Fujisawa R, Polo Rivera C, Labib KPM. Multiple UBX proteins reduce the ubiquitin threshold of the mammalian p97-UFD1-NPL4 unfoldase. eLife 2022; 11:e76763. [PMID: 35920641 PMCID: PMC9377798 DOI: 10.7554/elife.76763] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
The p97/Cdc48 ATPase and its ubiquitin receptors Ufd1-Npl4 are essential to unfold ubiquitylated proteins in many areas of eukaryotic cell biology. In yeast, Cdc48-Ufd1-Npl4 is controlled by a quality control mechanism, whereby substrates must be conjugated to at least five ubiquitins. Here, we show that mammalian p97-UFD1-NPL4 is governed by a complex interplay between additional p97 cofactors and the number of conjugated ubiquitins. Using reconstituted assays for the disassembly of ubiquitylated CMG (Cdc45-MCM-GINS) helicase by human p97-UFD1-NPL4, we show that the unfoldase has a high ubiquitin threshold for substrate unfolding, which can be reduced by the UBX proteins UBXN7, FAF1, or FAF2. Our data indicate that the UBX proteins function by binding to p97-UFD1-NPL4 and stabilising productive interactions between UFD1-NPL4 and K48-linked chains of at least five ubiquitins. Stimulation by UBXN7 is dependent upon known ubiquitin-binding motifs, whereas FAF1 and FAF2 use a previously uncharacterised coiled-coil domain to reduce the ubiquitin threshold of p97-UFD1-NPL4. We show that deleting the Ubnx7 and Faf1 genes impairs CMG disassembly during S-phase and mitosis and sensitises cells to reduced ubiquitin ligase activity. These findings indicate that multiple UBX proteins are important for the efficient unfolding of ubiquitylated proteins by p97-UFD1-NPL4 in mammalian cells.
Collapse
Affiliation(s)
- Ryo Fujisawa
- The MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Cristian Polo Rivera
- The MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| | - Karim PM Labib
- The MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of DundeeDundeeUnited Kingdom
| |
Collapse
|
15
|
Briata P, Caputo L, Zapparoli E, Marcaccini E, Passalacqua M, Brondolo L, Bordo D, Rossi A, Nicoletti C, Bucci G, Puri PL, Inga A, Gherzi R. LncRNA EPR-induced METTL7A1 modulates target gene translation. Nucleic Acids Res 2022; 50:7608-7622. [PMID: 35748870 PMCID: PMC9303270 DOI: 10.1093/nar/gkac544] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/24/2022] [Accepted: 06/09/2022] [Indexed: 12/23/2022] Open
Abstract
EPR is a long non-coding RNA (lncRNA) that controls cell proliferation in mammary gland cells by regulating gene transcription. Here, we report on Mettl7a1 as a direct target of EPR. We show that EPR induces Mettl7a1 transcription by rewiring three-dimensional chromatin interactions at the Mettl7a1 locus. Our data indicate that METTL7A1 contributes to EPR-dependent inhibition of TGF-β signaling. METTL7A1 is absent in tumorigenic murine mammary gland cells and its human ortholog (METTL7A) is downregulated in breast cancers. Importantly, re-expression of METTL7A1 in 4T1 tumorigenic cells attenuates their transformation potential, with the putative methyltransferase activity of METTL7A1 being dispensable for its biological functions. We found that METTL7A1 localizes in the cytoplasm whereby it interacts with factors implicated in the early steps of mRNA translation, associates with ribosomes, and affects the levels of target proteins without altering mRNA abundance. Overall, our data indicates that METTL7A1-a transcriptional target of EPR-modulates translation of select transcripts.
Collapse
Affiliation(s)
- Paola Briata
- Correspondence may also be addressed to Paola Briata. Tel: +39 010555540;
| | | | - Ettore Zapparoli
- Center for Omics Sciences, IRCCS Ospedale, San Raffaele, 20132 Milano, Italy
| | - Elisa Marcaccini
- Gene Expression Regulation Laboratory, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy,Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (3R Center), 56122 Pisa, Italy
| | - Lorenzo Brondolo
- Gene Expression Regulation Laboratory, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Domenico Bordo
- Gene Expression Regulation Laboratory, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Annalisa Rossi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Chiara Nicoletti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Gabriele Bucci
- Center for Omics Sciences, IRCCS Ospedale, San Raffaele, 20132 Milano, Italy
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | | | - Roberto Gherzi
- To whom correspondence should be addressed. Tel: +39 010555402; Emails: ;
| |
Collapse
|
16
|
Sar1 Affects the Localization of Perilipin 2 to Lipid Droplets. Int J Mol Sci 2022; 23:ijms23126366. [PMID: 35742827 PMCID: PMC9223735 DOI: 10.3390/ijms23126366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/03/2022] [Accepted: 06/03/2022] [Indexed: 02/05/2023] Open
Abstract
Lipid droplets (LDs) are intracellular organelles that are ubiquitous in many types of cells. The LD core consists of triacylglycerols (TGs) surrounded by a phospholipid monolayer and surface proteins such as perilipin 2 (PLIN2). Although TGs accumulate in the phospholipid bilayer of the endoplasmic reticulum (ER) and subsequently nascent LDs buds from ER, the mechanism by which LD proteins are transported to LD particles is not fully understood. Sar1 is a GTPase known as a regulator of coat protein complex Ⅱ (COPⅡ) vesicle budding, and its role in LD formation was investigated in this study. HuH7 human hepatoma cells were infected with adenoviral particles containing genes coding GFP fused with wild-type Sar1 (Sar1 WT) or a GTPase mutant form (Sar1 H79G). When HuH7 cells were treated with oleic acid, Sar1 WT formed a ring-like structure around the LDs. The transient expression of Sar1 did not significantly alter the levels of TG and PLIN2 in the cells. However, the localization of PLIN2 to the LDs decreased in the cells expressing Sar1 H79G. Furthermore, the effects of Sar1 on PLIN2 localization to the LDs were verified by the suppression of endogenous Sar1 using the short hairpin RNA technique. In conclusion, it was found that Sar1 has some roles in the intracellular distribution of PLIN2 to LDs in liver cells.
Collapse
|
17
|
Lyschik S, Lauer AA, Roth T, Janitschke D, Hollander M, Will T, Hartmann T, Kopito RR, Helms V, Grimm MOW, Schrul B. PEX19 Coordinates Neutral Lipid Storage in Cells in a Peroxisome-Independent Fashion. Front Cell Dev Biol 2022; 10:859052. [PMID: 35557938 PMCID: PMC9086359 DOI: 10.3389/fcell.2022.859052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Cellular lipid metabolism is tightly regulated and requires a sophisticated interplay of multiple subcellular organelles to adapt to changing nutrient supply. PEX19 was originally described as an essential peroxisome biogenesis factor that selectively targets membrane proteins to peroxisomes. Metabolic aberrations that were associated with compromised PEX19 functions, were solely attributed to the absence of peroxisomes, which is also considered the underlying cause for Zellweger Spectrum Disorders. More recently, however, it was shown that PEX19 also mediates the targeting of the VCP/P97-recuitment factor UBXD8 to the ER from where it partitions to lipid droplets (LDs) but the physiological consequences remained elusive. Here, we addressed the intriguing possibility that PEX19 coordinates the functions of the major cellular sites of lipid metabolism. We exploited the farnesylation of PEX19 and deciphered the organelle-specific functions of PEX19 using systems level approaches. Non-farnesylated PEX19 is sufficient to fully restore the metabolic activity of peroxisomes, while farnesylated PEX19 controls lipid metabolism by a peroxisome-independent mechanism that can be attributed to sorting a specific protein subset to LDs. In the absence of this PEX19-dependent LD proteome, cells accumulate excess triacylglycerols and fail to fully deplete their neutral lipid stores under catabolic conditions, highlighting a hitherto unrecognized function of PEX19 in controlling neutral lipid storage and LD dynamics.
Collapse
Affiliation(s)
- Sven Lyschik
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling (PZMS), Faculty of Medicine, Saarland University, Homburg, Germany
| | - Anna A. Lauer
- Experimental Neurology, Saarland University, Homburg, Germany
| | - Tanja Roth
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling (PZMS), Faculty of Medicine, Saarland University, Homburg, Germany
| | | | - Markus Hollander
- Center for Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Thorsten Will
- Center for Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Tobias Hartmann
- Experimental Neurology, Saarland University, Homburg, Germany
- Deutsches Institut für Demenzprävention, Saarland University, Homburg, Germany
| | - Ron R. Kopito
- Department of Biology, Stanford University, Stanford, CA, United States
| | - Volkhard Helms
- Center for Bioinformatics, Saarland University, Saarbruecken, Germany
| | - Marcus O. W. Grimm
- Experimental Neurology, Saarland University, Homburg, Germany
- Deutsches Institut für Demenzprävention, Saarland University, Homburg, Germany
- Nutrition Therapy and Counseling, Campus Rheinland, SRH University of Applied Health Sciences, Leverkusen, Germany
| | - Bianca Schrul
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling (PZMS), Faculty of Medicine, Saarland University, Homburg, Germany
- *Correspondence: Bianca Schrul,
| |
Collapse
|
18
|
Leznicki P, Schneider HO, Harvey JV, Shi WQ, High S. Co-translational biogenesis of lipid droplet integral membrane proteins. J Cell Sci 2022; 135:272279. [PMID: 34558621 PMCID: PMC8627552 DOI: 10.1242/jcs.259220] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/16/2021] [Indexed: 12/18/2022] Open
Abstract
Membrane proteins destined for lipid droplets (LDs), a major intracellular storage site for neutral lipids, are inserted into the endoplasmic reticulum (ER) and then trafficked to LDs where they reside in a hairpin loop conformation. Here, we show that LD membrane proteins can be delivered to the ER either co- or post-translationally and that their membrane-embedded region specifies pathway selection. The co-translational route for LD membrane protein biogenesis is insensitive to a small molecule inhibitor of the Sec61 translocon, Ipomoeassin F, and instead relies on the ER membrane protein complex (EMC) for membrane insertion. This route may even result in a transient exposure of the short N termini of some LD membrane proteins to the ER lumen, followed by putative topological rearrangements that would enable their transmembrane segment to form a hairpin loop and N termini to face the cytosol. Our study reveals an unexpected complexity to LD membrane protein biogenesis and identifies a role for the EMC during their co-translational insertion into the ER. Summary: Insertion of many lipid droplet membrane proteins into the ER is co-translational, mediated by the ER membrane protein complex and may involve topology reorientation.
Collapse
Affiliation(s)
- Pawel Leznicki
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | | | - Jada V Harvey
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Wei Q Shi
- Department of Chemistry, Ball State University, Muncie, IN 47306, USA
| | - Stephen High
- School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
19
|
Liao PC, Yang EJ, Borgman T, Boldogh IR, Sing CN, Swayne TC, Pon LA. Touch and Go: Membrane Contact Sites Between Lipid Droplets and Other Organelles. Front Cell Dev Biol 2022; 10:852021. [PMID: 35281095 PMCID: PMC8908909 DOI: 10.3389/fcell.2022.852021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/03/2022] [Indexed: 12/26/2022] Open
Abstract
Lipid droplets (LDs) have emerged not just as storage sites for lipids but as central regulators of metabolism and organelle quality control. These critical functions are achieved, in part, at membrane contact sites (MCS) between LDs and other organelles. MCS are sites of transfer of cellular constituents to or from LDs for energy mobilization in response to nutrient limitations, as well as LD biogenesis, expansion and autophagy. Here, we describe recent findings on the mechanisms underlying the formation and function of MCS between LDs and mitochondria, ER and lysosomes/vacuoles and the role of the cytoskeleton in promoting LD MCS through its function in LD movement and distribution in response to environmental cues.
Collapse
Affiliation(s)
- Pin-Chao Liao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Emily J. Yang
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Taylor Borgman
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States
| | - Istvan R. Boldogh
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Cierra N. Sing
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States
| | - Theresa C. Swayne
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Liza A. Pon
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States
- *Correspondence: Liza A. Pon,
| |
Collapse
|
20
|
Olarte MJ, Swanson JMJ, Walther TC, Farese RV. The CYTOLD and ERTOLD pathways for lipid droplet-protein targeting. Trends Biochem Sci 2022; 47:39-51. [PMID: 34583871 PMCID: PMC8688270 DOI: 10.1016/j.tibs.2021.08.007] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/23/2021] [Accepted: 08/24/2021] [Indexed: 01/03/2023]
Abstract
Lipid droplets (LDs) are the main organelles for lipid storage, and their surfaces contain unique proteins with diverse functions, including those that facilitate the deposition and mobilization of LD lipids. Among organelles, LDs have an unusual structure with an organic, hydrophobic oil phase covered by a phospholipid monolayer. The unique properties of LD monolayer surfaces require proteins to localize to LDs by distinct mechanisms. Here we review the two pathways known to mediate direct LD protein localization: the CYTOLD pathway mediates protein targeting from the cytosol toLDs, and the ERTOLD pathway functions in protein targeting from the endoplasmic reticulum toLDs. We describe the emerging principles for each targeting pathway in animal cells and highlight open questions in the field.
Collapse
Affiliation(s)
- Maria-Jesus Olarte
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | | | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02124, USA.
| | - Robert V Farese
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02124, USA.
| |
Collapse
|
21
|
Wong JM, Eirin-Lopez JM. Evolution of methyltransferase like (METTL) proteins in Metazoa: A complex gene family involved in epitranscriptomic regulation and other epigenetic processes. Mol Biol Evol 2021; 38:5309-5327. [PMID: 34480573 PMCID: PMC8662637 DOI: 10.1093/molbev/msab267] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The methyltransferase-like (METTL) proteins constitute a family of seven-beta-strand methyltransferases with S-adenosyl methionine-binding domains that modify DNA, RNA, and proteins. Methylation by METTL proteins contributes to the epigenetic, and in the case of RNA modifications, epitranscriptomic regulation of a variety of biological processes. Despite their functional importance, most investigations of the substrates and functions of METTLs within metazoans have been restricted to model vertebrate taxa. In the present work, we explore the evolutionary mechanisms driving the diversification and functional differentiation of 33 individual METTL proteins across Metazoa. Our results show that METTLs are nearly ubiquitous across the animal kingdom, with most having arisen early in metazoan evolution (i.e., occur in basal metazoan phyla). Individual METTL lineages each originated from single independent ancestors, constituting monophyletic clades, which suggests that each METTL was subject to strong selective constraints driving its structural and/or functional specialization. Interestingly, a similar process did not extend to the differentiation of nucleoside-modifying and protein-modifying METTLs (i.e., each METTL type did not form a unique monophyletic clade). The members of these two types of METTLs also exhibited differences in their rates of evolution. Overall, we provide evidence that the long-term evolution of METTL family members was driven by strong purifying selection, which in combination with adaptive selection episodes, led to the functional specialization of individual METTL lineages. This work contributes useful information regarding the evolution of a gene family that fulfills a variety of epigenetic functions, and can have profound influences on molecular processes and phenotypic traits.
Collapse
Affiliation(s)
- Juliet M Wong
- Environmental Epigenetics Laboratory, Institute of Environment, Florida International University, Miami, FL, United States
| | - Jose M Eirin-Lopez
- Environmental Epigenetics Laboratory, Institute of Environment, Florida International University, Miami, FL, United States
| |
Collapse
|
22
|
Campeanu IJ, Jiang Y, Liu L, Pilecki M, Najor A, Cobani E, Manning M, Zhang XM, Yang ZQ. Multi-omics integration of methyltransferase-like protein family reveals clinical outcomes and functional signatures in human cancer. Sci Rep 2021; 11:14784. [PMID: 34285249 PMCID: PMC8292347 DOI: 10.1038/s41598-021-94019-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/04/2021] [Indexed: 01/13/2023] Open
Abstract
Human methyltransferase-like (METTL) proteins transfer methyl groups to nucleic acids, proteins, lipids, and other small molecules, subsequently playing important roles in various cellular processes. In this study, we performed integrated genomic, transcriptomic, proteomic, and clinicopathological analyses of 34 METTLs in a large cohort of primary tumor and cell line data. We identified a subset of METTL genes, notably METTL1, METTL7B, and NTMT1, with high frequencies of genomic amplification and/or up-regulation at both the mRNA and protein levels in a spectrum of human cancers. Higher METTL1 expression was associated with high-grade tumors and poor disease prognosis. Loss-of-function analysis in tumor cell lines indicated the biological importance of METTL1, an m7G methyltransferase, in cancer cell growth and survival. Furthermore, functional annotation and pathway analysis of METTL1-associated proteins revealed that, in addition to the METTL1 cofactor WDR4, RNA regulators and DNA packaging complexes may be functionally interconnected with METTL1 in human cancer. Finally, we generated a crystal structure model of the METTL1–WDR4 heterodimeric complex that might aid in understanding the key functional residues. Our results provide new information for further functional study of some METTL alterations in human cancer and might lead to the development of small inhibitors that target cancer-promoting METTLs.
Collapse
Affiliation(s)
- Ion John Campeanu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yuanyuan Jiang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Lanxin Liu
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Maksymilian Pilecki
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Alvina Najor
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Era Cobani
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Morenci Manning
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Xiaohong Mary Zhang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, 4100 John R Street, HWCRC 815, Detroit, MI, 48201, USA
| | - Zeng-Quan Yang
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA. .,Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, 4100 John R Street, HWCRC 815, Detroit, MI, 48201, USA.
| |
Collapse
|
23
|
Methyltransferase-like protein 7A (METTL7A) promotes cell survival and osteogenic differentiation under metabolic stress. Cell Death Discov 2021; 7:154. [PMID: 34226523 PMCID: PMC8257615 DOI: 10.1038/s41420-021-00555-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/07/2021] [Accepted: 05/29/2021] [Indexed: 12/24/2022] Open
Abstract
While bone has an inherent capacity to heal itself, it is very difficult to reconstitute large bone defects. Regenerative medicine, including stem cell implantation, has been studied as a novel solution to treat these conditions. However, when the local vascularity is impaired, even the transplanted cells undergo rapid necrosis before differentiating into osteoblasts and regenerating bone. Thus, to increase the effectiveness of stem cell transplantation, it is quintessential to improve the viability of the implanted stem cells. In this study, given that the regulation of glucose may hold the key to stem cell survival and osteogenic differentiation, we investigated the molecules that can replace the effect of glucose under ischemic microenvironment of stem cell transplantation in large bone defects. By analyzing differentially expressed genes under glucose-supplemented and glucose-free conditions, we explored markers such as methyltransferase-like protein 7A (METTL7A) that are potentially related to cell survival and osteogenic differentiation. Overexpression of METTL7A gene enhanced the osteogenic differentiation and viability of human bone marrow stem cells (hBMSCs) in glucose-free conditions. When the in vivo effectiveness of METTL7A-transfected cells in bone regeneration was explored in a rat model of critical-size segmental long-bone defect, METTL7A-transfected hBMSCs showed significantly better regenerative potential than the control vector-transfected hBMSCs. DNA methylation profiles showed a large difference in methylation status of genes related to osteogenesis and cell survival between hBMSCs cultured in glucose-supplemented condition and those cultured in glucose-free condition. Interestingly, METTL7A overexpression altered the methylation status of related genes to favor osteogenic differentiation and cell survival. In conclusion, it is suggested that a novel factor METTL7A enhances osteogenic differentiation and viability of hBMSCs by regulating the methylation status of genes related to osteogenesis or survival.
Collapse
|
24
|
Gwon Y, Maxwell BA, Kolaitis RM, Zhang P, Kim HJ, Taylor JP. Ubiquitination of G3BP1 mediates stress granule disassembly in a context-specific manner. Science 2021; 372:eabf6548. [PMID: 34739333 PMCID: PMC8574224 DOI: 10.1126/science.abf6548] [Citation(s) in RCA: 192] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Stress granules are dynamic, reversible condensates composed of RNA and protein that assemble in eukaryotic cells in response to a variety of stressors and are normally disassembled after stress is removed. The composition and assembly of stress granules is well understood, but little is known about the mechanisms that govern disassembly. Impaired disassembly has been implicated in some diseases including amyotrophic lateral sclerosis, frontotemporal dementia, and multisystem proteinopathy. Using cultured human cells, we found that stress granule disassembly was context-dependent: Specifically in the setting of heat shock, disassembly required ubiquitination of G3BP1, the central protein within the stress granule RNA-protein network. We found that ubiquitinated G3BP1 interacted with the endoplasmic reticulum–associated protein FAF2, which engaged the ubiquitin-dependent segregase p97/VCP (valosin-containing protein). Thus, targeting of G3BP1 weakened the stress granule–specific interaction network, resulting in granule disassembly.
Collapse
Affiliation(s)
- Youngdae Gwon
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Brian A. Maxwell
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Regina-Maria Kolaitis
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Peipei Zhang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Hong Joo Kim
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - J. Paul Taylor
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815
| |
Collapse
|
25
|
Chen HK, Rosset SL, Wang LH, Chen CS. The characteristics of host lipid body biogenesis during coral-dinoflagellate endosymbiosis. PeerJ 2021; 9:e11652. [PMID: 34221732 PMCID: PMC8234918 DOI: 10.7717/peerj.11652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/31/2021] [Indexed: 11/25/2022] Open
Abstract
Intracellular lipid body (LB) biogenesis depends on the symbiosis between coral hosts and their Symbiodinaceae. Therefore, understanding the mechanism(s) behind LB biosynthesis in corals can portentially elucide the drivers of cellular regulation during endosymbiosis. This study assessed LB formation in the gastrodermal tissue layer of the hermatypic coral Euphyllia glabrescens. Diel rhythmicity in LB size and distribution was observed; solar irradiation onset at sunrise initiated an increase in LB formation, which continued throughout the day and peaked after sunset at 18:00. The LBs migrated from the area near the mesoglea to the gastrodermal cell border near the coelenteron. Micro-LB biogenesis occurred in the endoplasmic reticulum (ER) of the host gastrodermal cells. A transcriptomic analysis of genes related to lipogenesis indicated that binding immunoglobulin protein (BiP) plays a key role in metabolic signaling pathways. The diel rhythmicity of LB biogenesis was correlated with ER-localized BiP expression. BiP expression peaked during the period with the largest increase in LB formation, thereby indicating that the chaperoning reaction of abnormal protein folding inside the host ER is likely involved in LB biosynthesis. These findings suggest that the host ER, central to LB formation, potentially facilitates the regulation of endosymbiosis between coral hosts and Symbiodiniaceae.
Collapse
Affiliation(s)
- Hung-Kai Chen
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan
| | - Sabrina L Rosset
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan
| | - Li-Hsueh Wang
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan.,Graduate Institute of Marine Biology, National Dong-Hwa University, Pingtung, Taiwan
| | - Chii-Shiarng Chen
- National Museum of Marine Biology and Aquarium, Pingtung, Taiwan.,Graduate Institute of Marine Biology, National Dong-Hwa University, Pingtung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
26
|
Khaddaj R, Mari M, Cottier S, Reggiori F, Schneiter R. The surface of lipid droplets constitutes a barrier for endoplasmic reticulum-resident integral membrane proteins. J Cell Sci 2021; 135:268334. [PMID: 34028531 DOI: 10.1242/jcs.256206] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/14/2021] [Indexed: 12/12/2022] Open
Abstract
Lipid droplets (LDs) are globular subcellular structures that store neutral lipids. LDs are closely associated with the endoplasmic reticulum (ER) and are limited by a phospholipid monolayer harboring a specific set of proteins. Most of these proteins associate with LDs through either an amphipathic helix or a membrane-embedded hairpin motif. Here, we address the question of whether integral membrane proteins can localize to the surface of LDs. To test this, we fused perilipin 3 (PLIN3), a mammalian LD-targeted protein, to ER-resident proteins. The resulting fusion proteins localized to the periphery of LDs in both yeast and mammalian cells. This peripheral LD localization of the fusion proteins, however, was due to a redistribution of the ER around LDs, as revealed by bimolecular fluorescence complementation between ER- and LD-localized partners. A LD-tethering function of PLIN3-containing membrane proteins was confirmed by fusing PLIN3 to the cytoplasmic domain of an outer mitochondrial membrane protein, OM14. Expression of OM14-PLIN3 induced a close apposition between LDs and mitochondria. These data indicate that the ER-LD junction constitutes a barrier for ER-resident integral membrane proteins.
Collapse
Affiliation(s)
- Rasha Khaddaj
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Muriel Mari
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Stéphanie Cottier
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells and Systems, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
27
|
Wang L, Liu J, Miao Z, Pan Q, Cao W. Lipid droplets and their interactions with other organelles in liver diseases. Int J Biochem Cell Biol 2021; 133:105937. [PMID: 33529713 DOI: 10.1016/j.biocel.2021.105937] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/07/2020] [Accepted: 01/06/2021] [Indexed: 02/01/2023]
Abstract
Lipid droplets are cellular organelles used for lipid storage with a hydrophobic core of neutral lipids enclosed by a phospholipid monolayer. Besides presenting as giant single organelles in fat tissue, lipid droplets are also widely present as a multitude of small structures in hepatocytes, where they play key roles in health and disease of the liver. In addition to lipid storage, lipid droplets are also directly involved in lipid metabolism, membrane biosynthesis, cell signaling, inflammation, pathogen-host interaction and cancer development. In addition, they interact with other cellular organelles to regulate cellular biology. It is fair to say that the exact functions of lipid droplets in cellular physiology remain largely obscure. Thus prompted, here we aim to analyze the corpus of contemporary biomedical literature to create a framework as to how the role of lipid droplets in hepatocyte physiology and pathophysiology should be understood. The resulting framework should help understanding the interaction of lipid droplets with other organelles in important liver diseases, including fatty liver disease, viral hepatitis and liver cancer and direct further research directions.
Collapse
Affiliation(s)
- Ling Wang
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Jiaye Liu
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Zhijiang Miao
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands.
| | - Wanlu Cao
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
28
|
Lopata A, Kniss A, Löhr F, Rogov VV, Dötsch V. Ubiquitination in the ERAD Process. Int J Mol Sci 2020; 21:ijms21155369. [PMID: 32731622 PMCID: PMC7432864 DOI: 10.3390/ijms21155369] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 01/08/2023] Open
Abstract
In this review, we focus on the ubiquitination process within the endoplasmic reticulum associated protein degradation (ERAD) pathway. Approximately one third of all synthesized proteins in a cell are channeled into the endoplasmic reticulum (ER) lumen or are incorporated into the ER membrane. Since all newly synthesized proteins enter the ER in an unfolded manner, folding must occur within the ER lumen or co-translationally, rendering misfolding events a serious threat. To prevent the accumulation of misfolded protein in the ER, proteins that fail the quality control undergo retrotranslocation into the cytosol where they proceed with ubiquitination and degradation. The wide variety of misfolded targets requires on the one hand a promiscuity of the ubiquitination process and on the other hand a fast and highly processive mechanism. We present the various ERAD components involved in the ubiquitination process including the different E2 conjugating enzymes, E3 ligases, and E4 factors. The resulting K48-linked and K11-linked ubiquitin chains do not only represent a signal for degradation by the proteasome but are also recognized by the AAA+ ATPase Cdc48 and get in the process of retrotranslocation modified by enzymes bound to Cdc48. Lastly we discuss the conformations adopted in particular by K48-linked ubiquitin chains and their importance for degradation.
Collapse
|
29
|
Dhiman R, Caesar S, Thiam AR, Schrul B. Mechanisms of protein targeting to lipid droplets: A unified cell biological and biophysical perspective. Semin Cell Dev Biol 2020; 108:4-13. [PMID: 32201131 DOI: 10.1016/j.semcdb.2020.03.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 02/23/2020] [Accepted: 03/13/2020] [Indexed: 12/22/2022]
Abstract
Lipid droplets (LDs), or oil bodies in plants, are specialized organelles that primarily serve as hubs of cellular metabolic energy storage and consumption. These ubiquitous cytoplasmic organelles are derived from the endoplasmic reticulum (ER) and consist of a hydrophobic neutral lipid core - mainly consisting of triglycerides and sterol esters - that is encircled by a phospholipid monolayer. The dynamic metabolic functions of the LDs are mainly executed and regulated by proteins on the monolayer surface. However, its unique architecture puts some structural constraints on the types of proteins that can associate with LDs. The lipid monolayer is decorated with either peripheral proteins or with integral membrane proteins that adopt a monotopic topology. Due to its oil-water interface, which is energetically costly, the LD surface happens to be favorable to the recruitment of many proteins involved in metabolic but also non-metabolic functions. We only started very recently to understand biophysical and biochemical principles controlling protein targeting to LDs. This review aims to summarize the most recent findings regarding this topic and proposes directions that will potentially lead to a better understanding of LD surface characteristics, as compared to bilayer membranes, and how that impacts protein-LD interactions.
Collapse
Affiliation(s)
- Ravi Dhiman
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling (PZMS), Faculty of Medicine, Saarland University, 66421 Homburg, Saar, Germany
| | - Stefanie Caesar
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling (PZMS), Faculty of Medicine, Saarland University, 66421 Homburg, Saar, Germany
| | - Abdou Rachid Thiam
- Laboratoire de Physique de l'École Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Université de Paris, F-75005 Paris, France.
| | - Bianca Schrul
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling (PZMS), Faculty of Medicine, Saarland University, 66421 Homburg, Saar, Germany.
| |
Collapse
|
30
|
Exner T, Romero-Brey I, Yifrach E, Rivera-Monroy J, Schrul B, Zouboulis CC, Stremmel W, Honsho M, Bartenschlager R, Zalckvar E, Poppelreuther M, Füllekrug J. An alternative membrane topology permits lipid droplet localization of peroxisomal fatty acyl-CoA reductase 1. J Cell Sci 2019; 132:jcs.223016. [PMID: 30745342 DOI: 10.1242/jcs.223016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 02/01/2019] [Indexed: 01/02/2023] Open
Abstract
Fatty acyl-CoA reductase 1 (Far1) is a ubiquitously expressed peroxisomal membrane protein that generates the fatty alcohols required for the biosynthesis of ether lipids. Lipid droplet localization of exogenously expressed and endogenous human Far1 was observed by fluorescence microscopy under conditions of increased triglyceride synthesis in tissue culture cells. This unexpected finding was supported further by correlative light electron microscopy and subcellular fractionation. Selective permeabilization, protease sensitivity and N-glycosylation tagging suggested that Far1 is able to assume two different membrane topologies, differing in the orientation of the short hydrophilic C-terminus towards the lumen or the cytosol, respectively. Two closely spaced hydrophobic domains are contained within the C-terminal region. When analyzed separately, the second domain was sufficient for the localization of a fluorescent reporter to lipid droplets. Targeting of Far1 to lipid droplets was not impaired in either Pex19 or ASNA1 (also known as TRC40) CRISPR/Cas9 knockout cells. In conclusion, our data suggest that Far1 is a novel member of the rather exclusive group of dual topology membrane proteins. At the same time, Far1 shows lipid metabolism-dependent differential subcellular localizations to peroxisomes and lipid droplets.
Collapse
Affiliation(s)
- Tarik Exner
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, 69120 Heidelberg, Germany
| | - Inés Romero-Brey
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Eden Yifrach
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Jhon Rivera-Monroy
- Department of Molecular Biology, University Medical Center Göttingen, 37077 Göttingen, Germany
| | - Bianca Schrul
- Medical Biochemistry and Molecular Biology, Center for Molecular Signaling (PZMS), Saarland University, 66421 Homburg/Saar, Germany
| | - Christos C Zouboulis
- Departments of Dermatology, Venereology, Allergology and Immunology, Dessau Medical Center, Brandenburg Medical School Theodor Fontane, 06847 Dessau, Germany
| | - Wolfgang Stremmel
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, 69120 Heidelberg, Germany
| | - Masanori Honsho
- Medical Institute of Bioregulation, Kyushu University, 812-8582 Fukuoka, Japan
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Einat Zalckvar
- Department of Molecular Genetics, Weizmann Institute of Science, 7610001 Rehovot, Israel
| | - Margarete Poppelreuther
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, 69120 Heidelberg, Germany
| | - Joachim Füllekrug
- Molecular Cell Biology Laboratory Internal Medicine IV, University of Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
31
|
Vieyres G, Pietschmann T. HCV Pit Stop at the Lipid Droplet: Refuel Lipids and Put on a Lipoprotein Coat before Exit. Cells 2019; 8:cells8030233. [PMID: 30871009 PMCID: PMC6468556 DOI: 10.3390/cells8030233] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 02/28/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023] Open
Abstract
The replication cycle of the liver-tropic hepatitis C virus (HCV) is tightly connected to the host lipid metabolism, during the virus entry, replication, assembly and egress stages, but also while the virus circulates in the bloodstream. This interplay coins viral particle properties, governs viral cell tropism, and facilitates immune evasion. This review summarizes our knowledge of these interactions focusing on the late steps of the virus replication cycle. It builds on our understanding of the cell biology of lipid droplets and the biosynthesis of liver lipoproteins and attempts to explain how HCV hijacks these organelles and pathways to assemble its lipo-viro-particles. In particular, this review describes (i) the mechanisms of viral protein translocation to and from the lipid droplet surface and the orchestration of an interface between replication and assembly complexes, (ii) the importance of the triglyceride mobilization from the lipid droplets for HCV assembly, (iii) the interplay between HCV and the lipoprotein synthesis pathway including the role played by apolipoproteins in virion assembly, and finally (iv) the consequences of these complex virus–host interactions on the virion composition and its biophysical properties. The wealth of data accumulated in the past years on the role of the lipid metabolism in HCV assembly and its imprint on the virion properties will guide vaccine design efforts and reinforce our understanding of the hepatic lipid metabolism in health and disease.
Collapse
Affiliation(s)
- Gabrielle Vieyres
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), 30625 Hannover, Germany.
| | - Thomas Pietschmann
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Centre for Infection Research (HZI), 30625 Hannover, Germany.
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 38124 Braunschweig, Germany.
| |
Collapse
|
32
|
Krahmer N, Najafi B, Schueder F, Quagliarini F, Steger M, Seitz S, Kasper R, Salinas F, Cox J, Uhlenhaut NH, Walther TC, Jungmann R, Zeigerer A, Borner GHH, Mann M. Organellar Proteomics and Phospho-Proteomics Reveal Subcellular Reorganization in Diet-Induced Hepatic Steatosis. Dev Cell 2018; 47:205-221.e7. [PMID: 30352176 DOI: 10.1016/j.devcel.2018.09.017] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 06/29/2018] [Accepted: 09/18/2018] [Indexed: 01/20/2023]
Abstract
Lipid metabolism is highly compartmentalized between cellular organelles that dynamically adapt their compositions and interactions in response to metabolic challenges. Here, we investigate how diet-induced hepatic lipid accumulation, observed in non-alcoholic fatty liver disease (NAFLD), affects protein localization, organelle organization, and protein phosphorylation in vivo. We develop a mass spectrometric workflow for protein and phosphopeptide correlation profiling to monitor levels and cellular distributions of ∼6,000 liver proteins and ∼16,000 phosphopeptides during development of steatosis. Several organelle contact site proteins are targeted to lipid droplets (LDs) in steatotic liver, tethering organelles orchestrating lipid metabolism. Proteins of the secretory pathway dramatically redistribute, including the mis-localization of the COPI complex and sequestration of the Golgi apparatus at LDs. This correlates with reduced hepatic protein secretion. Our systematic in vivo analysis of subcellular rearrangements and organelle-specific phosphorylation reveals how nutrient overload leads to organellar reorganization and cellular dysfunction.
Collapse
Affiliation(s)
- Natalie Krahmer
- Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Bahar Najafi
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg 85764, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - Florian Schueder
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany; Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Fabiana Quagliarini
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), IDO, Garching, Munich 85748, Germany
| | - Martin Steger
- Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Susanne Seitz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg 85764, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - Robert Kasper
- Max Planck Institute of Neurobiology, Imaging facility, Martinsried 82152, Germany
| | - Favio Salinas
- Computational Systems Biochemistry, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Jürgen Cox
- Computational Systems Biochemistry, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Nina Henriette Uhlenhaut
- Helmholtz Diabetes Center (HMGU) and German Center for Diabetes Research (DZD), IDO, Garching, Munich 85748, Germany
| | - Tobias Christian Walther
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Ralf Jungmann
- Department of Physics and Center for Nanoscience, Ludwig Maximilian University, Munich, Germany; Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg 85764, Germany; Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg 85764, Germany
| | - Georg Heinz Helmut Borner
- Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Matthias Mann
- Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried 82152, Germany; Faculty of Health Sciences, NNF Center for Protein Research, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
33
|
Zhang C, Liu P. The New Face of the Lipid Droplet: Lipid Droplet Proteins. Proteomics 2018; 19:e1700223. [DOI: 10.1002/pmic.201700223] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/13/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Congyan Zhang
- National Laboratory of BiomacromoleculesCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of Sciences Beijing 100101 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Pingsheng Liu
- National Laboratory of BiomacromoleculesCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of Sciences Beijing 100101 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
34
|
Deruyffelaere C, Purkrtova Z, Bouchez I, Collet B, Cacas JL, Chardot T, Gallois JL, D'Andrea S. PUX10 Is a CDC48A Adaptor Protein That Regulates the Extraction of Ubiquitinated Oleosins from Seed Lipid Droplets in Arabidopsis. THE PLANT CELL 2018; 30:2116-2136. [PMID: 30087208 PMCID: PMC6181022 DOI: 10.1105/tpc.18.00275] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 06/06/2018] [Accepted: 07/31/2018] [Indexed: 05/19/2023]
Abstract
Postgerminative mobilization of neutral lipids stored in seed lipid droplets (LDs) is preceded by the degradation of oleosins, the major structural LD proteins that stabilize LDs in dry seeds. We previously showed that Arabidopsis thaliana oleosins are marked for degradation by ubiquitination and are extracted from LDs before proteolysis. However, the mechanisms underlying the dislocation of these LD-anchored proteins from the LD monolayer are yet unknown. Here, we report that PUX10, a member of the plant UBX-domain containing (PUX) protein family, is an integral LD protein that associates with a subpopulation of LDs during seed germination. In pux10 mutant seedlings, PUX10 deficiency impaired the degradation of ubiquitinated oleosins and prevented the extraction of ubiquitinated oleosins from LDs. We also showed that PUX10 interacts with ubiquitin and CDC48A, the AAA ATPase Cell Division Cycle 48, through its UBA and UBX domains, respectively. Collectively, these results strongly suggest that PUX10 is an adaptor recruiting CDC48A to ubiquitinated oleosins, thus facilitating the dislocation of oleosins from LDs by the segregase activity of CDC48A. We propose that PUX10 and CDC48A are core components of a LD-associated degradation machinery, which we named the LD-associated degradation system. Importantly, PUX10 is also the first determinant of a LD subpopulation described in plants, suggesting functional differentiation of LDs in Arabidopsis seedlings.
Collapse
Affiliation(s)
- Carine Deruyffelaere
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| | - Zita Purkrtova
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| | - Isabelle Bouchez
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| | - Boris Collet
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| | - Jean-Luc Cacas
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| | - Thierry Chardot
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| | | | - Sabine D'Andrea
- Institut Jean-Pierre Bourgin, INRA, AgroParisTech, CNRS, Université Paris-Saclay, 78000 Versailles, France
| |
Collapse
|
35
|
Proteomic analysis of monolayer-integrated proteins on lipid droplets identifies amphipathic interfacial α-helical membrane anchors. Proc Natl Acad Sci U S A 2018; 115:E8172-E8180. [PMID: 30104359 DOI: 10.1073/pnas.1807981115] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite not spanning phospholipid bilayers, monotopic integral proteins (MIPs) play critical roles in organizing biochemical reactions on membrane surfaces. Defining the structural basis by which these proteins are anchored to membranes has been hampered by the paucity of unambiguously identified MIPs and a lack of computational tools that accurately distinguish monolayer-integrating motifs from bilayer-spanning transmembrane domains (TMDs). We used quantitative proteomics and statistical modeling to identify 87 high-confidence candidate MIPs in lipid droplets, including 21 proteins with predicted TMDs that cannot be accommodated in these monolayer-enveloped organelles. Systematic cysteine-scanning mutagenesis showed the predicted TMD of one candidate MIP, DHRS3, to be a partially buried amphipathic α-helix in both lipid droplet monolayers and the cytoplasmic leaflet of endoplasmic reticulum membrane bilayers. Coarse-grained molecular dynamics simulations support these observations, suggesting that this helix is most stable at the solvent-membrane interface. The simulations also predicted similar interfacial amphipathic helices when applied to seven additional MIPs from our dataset. Our findings suggest that interfacial helices may be a common motif by which MIPs are integrated into membranes, and provide high-throughput methods to identify and study MIPs.
Collapse
|
36
|
Xu S, Zhang X, Liu P. Lipid droplet proteins and metabolic diseases. Biochim Biophys Acta Mol Basis Dis 2018; 1864:1968-1983. [DOI: 10.1016/j.bbadis.2017.07.019] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/14/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
|
37
|
Zhou S, Shen Y, Zheng M, Wang L, Che R, Hu W, Li P. DNA methylation of METTL7A gene body regulates its transcriptional level in thyroid cancer. Oncotarget 2018; 8:34652-34660. [PMID: 28416772 PMCID: PMC5470999 DOI: 10.18632/oncotarget.16147] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 02/08/2017] [Indexed: 11/25/2022] Open
Abstract
DNA methylation is the best-studied epigenetic mechanism for regulating gene transcription and maintaining genome stability. Current research progress of transcriptional regulation by DNA methylation mostly focuses on promoter region where hypomethylated CpG islands are present transcriptional activity, as hypermethylated CpG islands generally result in gene repression. Recently, the DNA methylation patterns across the gene body (intragenic methylation) have increasingly attracted attention towards their role in transcriptional regulation and efficiency, due to the improvement of numerous genome-wide DNA methylation profiling studies. However, the function and mechanism of gene body methylation is still unclear. In this study, we revealed that the methylation level of METTL7A, a seldom studied gene, was downregulated in thyroid cancer compared to normal thyroid cells in vivo and in vitro. Moreover, we determined the methylation level of one CpG site at the exon of the METTL7A gene body impacted the transcriptional activity. Through generating a mutation of this CpG site (CG to CC) of METTL7A exogenous vector artificially in vitro, we observed higher RNA polymerase II recruitment and a declined enrichment of methyl-CpG binding protein-2 in gene body of METTL7A, in papillary thryoid cancer cells (BCPAP) compared to normal thryoid cells. Finally, we revealed that EZH2, a subunit of polycomb repressor complex 2, dominant in thyroid cancer, might be responsible for regulating gene body methylation of METTL7A. Our study depicted the DNA methylation patterns and the transcriptional regulatory mechanism of the gene body in thyroid cancer. Furthermore, this study provides new insight into potential future avenues, for therapies targeting cancer.
Collapse
Affiliation(s)
- Shasha Zhou
- Department of Endocrinology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200040, People's Republic of China
| | - Yihang Shen
- Institute of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai, 200032, People's Republic of China
| | - Min Zheng
- Department of Oncology of Tangshan People's Hospital & Tangshan Cancer Hospital, North China University of Science and Technology, Tangshan, Hebei, 063001, People's Republic of China
| | - Linlin Wang
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, 250013, People's Republic of China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, Shandong, 250117, People's Republic of China
| | - Raymond Che
- University of Hawaii Cancer Center, University of Hawaii, Honolulu, HI, 96814, USA
| | - Wanning Hu
- Department of Oncology of Tangshan People's Hospital & Tangshan Cancer Hospital, North China University of Science and Technology, Tangshan, Hebei, 063001, People's Republic of China
| | - Pin Li
- Department of Endocrinology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, 200040, People's Republic of China
| |
Collapse
|
38
|
Inloes JM, Kiosses WB, Wang H, Walther TC, Farese RV, Cravatt BF. Functional Contribution of the Spastic Paraplegia-Related Triglyceride Hydrolase DDHD2 to the Formation and Content of Lipid Droplets. Biochemistry 2017; 57:827-838. [PMID: 29278326 DOI: 10.1021/acs.biochem.7b01028] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Deleterious mutations in the serine lipase DDHD2 are a causative basis of complex hereditary spastic paraplegia (HSP, subtype SPG54) in humans. We recently found that DDHD2 is a principal triglyceride hydrolase in the central nervous system (CNS) and that genetic deletion of this enzyme in mice leads to ectopic lipid droplet (LD) accumulation in neurons throughout the brain. Nonetheless, how HSP-related mutations in DDHD2 relate to triglyceride metabolism and LD formation remains poorly understood. Here, we have characterized a set of HSP-related mutations in DDHD2 and found that they disrupt triglyceride hydrolase activity in vitro and impair the capacity of DDHD2 to protect cells from LD accumulation following exposure to free fatty acid, an outcome that was also observed with a DDHD2-selective inhibitor. We furthermore isolated and characterized LDs from brain tissue of DDHD2-/- mice, revealing that they contain both established LD-associated proteins identified previously in other organs and CNS-enriched proteins, including several proteins with genetic links to human neurological disease. These data, taken together, indicate that the genetic inactivation of DDHD2, as caused by HSP-associated mutations, substantially perturbs lipid homeostasis and the formation and content of LDs, underscoring the importance of triglyceride metabolism for normal CNS function and the key role that DDHD2 plays in this process.
Collapse
Affiliation(s)
- Jordon M Inloes
- Department of Chemical Physiology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| | - William B Kiosses
- Department of Molecular Medicine, The Skaggs Institute for Chemical Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| | - Huajin Wang
- University Libraries, Carnegie Mellon University , Pittsburgh, Pennsylvania 15213, United States.,Department of Biological Sciences, Carnegie Mellon University , Pittsburgh, Pennsylvania 15213, United States.,Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health , Boston, Massachusetts 02115, United States.,Department of Cell Biology, Harvard Medical School , Boston, Massachusetts 02115, United States
| | - Tobias C Walther
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health , Boston, Massachusetts 02115, United States.,Department of Cell Biology, Harvard Medical School , Boston, Massachusetts 02115, United States.,Broad Institute of Harvard and MIT , Cambridge, Massachusetts 02142, United States.,Howard Hughes Medical Institute , Boston, Massachusetts 02115, United States
| | - Robert V Farese
- Department of Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health , Boston, Massachusetts 02115, United States.,Department of Cell Biology, Harvard Medical School , Boston, Massachusetts 02115, United States.,Broad Institute of Harvard and MIT , Cambridge, Massachusetts 02142, United States
| | - Benjamin F Cravatt
- Department of Chemical Physiology, The Skaggs Institute for Chemical Biology, The Scripps Research Institute , La Jolla, California 92037, United States
| |
Collapse
|
39
|
Loregger A, Raaben M, Tan J, Scheij S, Moeton M, van den Berg M, Gelberg-Etel H, Stickel E, Roitelman J, Brummelkamp T, Zelcer N. Haploid Mammalian Genetic Screen Identifies UBXD8 as a Key Determinant of HMGCR Degradation and Cholesterol Biosynthesis. Arterioscler Thromb Vasc Biol 2017; 37:2064-2074. [PMID: 28882874 PMCID: PMC5671778 DOI: 10.1161/atvbaha.117.310002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 08/29/2017] [Indexed: 01/23/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— The cellular demand for cholesterol requires control of its biosynthesis by the mevalonate pathway. Regulation of HMGCR (3-hydroxy-3-methylglutaryl coenzyme A reductase), a rate-limiting enzyme in this pathway and the target of statins, is a key control point herein. Accordingly, HMGCR is subject to negative and positive regulation. In particular, the ability of oxysterols and intermediates of the mevalonate pathway to stimulate its proteasomal degradation is an exquisite example of metabolically controlled feedback regulation. To define the genetic determinants that govern this process, we conducted an unbiased haploid mammalian genetic screen. Approach and Results— We generated human haploid cells with mNeon fused to endogenous HMGCR using CRISPR/Cas9 and used these cells to interrogate regulation of HMGCR abundance in live cells. This resulted in identification of known and new regulators of HMGCR, and among the latter, UBXD8 (ubiquitin regulatory X domain-containing protein 8), a gene that has not been previously implicated in this process. We demonstrate that UBXD8 is an essential determinant of metabolically stimulated degradation of HMGCR and of cholesterol biosynthesis in multiple cell types. Accordingly, UBXD8 ablation leads to aberrant cholesterol synthesis due to loss of feedback control. Mechanistically, we show that UBXD8 is necessary for sterol-stimulated dislocation of ubiquitylated HMGCR from the endoplasmic reticulum membrane en route to proteasomal degradation, a function dependent on its UBX domain. Conclusions— We establish UBXD8 as a previously unrecognized determinant that couples flux across the mevalonate pathway to control of cholesterol synthesis and demonstrate the feasibility of applying mammalian haploid genetics to study metabolic traits.
Collapse
Affiliation(s)
- Anke Loregger
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Matthijs Raaben
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Josephine Tan
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Saskia Scheij
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Martina Moeton
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Marlene van den Berg
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Hila Gelberg-Etel
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Elmer Stickel
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Joseph Roitelman
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Thijn Brummelkamp
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.)
| | - Noam Zelcer
- From the Department of Medical Biochemistry, Academic Medical Center of the University of Amsterdam, The Netherlands (A.L., J.T., S.S., M.M., M.v.d.B., N.Z.); Division of Biochemistry, The Netherlands Cancer Institute, Amsterdam (M.R., E.S., T.B.); CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna (T.B.); Cancer GenomiCs.nl, Amsterdam, The Netherlands (T.B.); Bert W. Strassburger Lipid Center, Sheba Medical Center, Tel Hashomer, Israel (H.G.-E., J.R.); and Department of Human Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Israel (H.G.-E., J.R.).
| |
Collapse
|
40
|
Barbosa AD, Siniossoglou S. Function of lipid droplet-organelle interactions in lipid homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1459-1468. [DOI: 10.1016/j.bbamcr.2017.04.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/31/2017] [Accepted: 04/02/2017] [Indexed: 12/20/2022]
|
41
|
Schuldiner M, Bohnert M. A different kind of love - lipid droplet contact sites. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28627434 DOI: 10.1016/j.bbalip.2017.06.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Lipid droplets (LDs) store lipids and hence serve as energy reservoir and as a source for building-blocks for the organelle membrane systems. LD biology therefore depends on tight communication with other organelles. The unique architecture of LDs, consisting of a neutral lipid core shielded by a phospholipid-monolayer, is however an obstacle to bulk-exchange of bilayer-bounded vesicles with other organelles. In recent years, it is emerging that contact sites, places where two organelles are positioned in close proximity allowing vesicle-independent communication, are an important way to integrate LDs into the organellar landscape. However, few LD contact sites have been studied in depth and our understanding of their structure, extent and function is only starting to emerge. Here, we highlight recent findings on the functions of LD contact sites and on the proteins involved in their formation and hypothesize about the unique characteristics of the contact sites formed by these intriguing organelles. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maria Bohnert
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
42
|
Bersuker K, Olzmann JA. Establishing the lipid droplet proteome: Mechanisms of lipid droplet protein targeting and degradation. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28627435 DOI: 10.1016/j.bbalip.2017.06.006] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Lipid droplets (LDs) are ubiquitous, endoplasmic reticulum (ER)-derived organelles that mediate the sequestration of neutral lipids (e.g. triacylglycerol and sterol esters), providing a dynamic cellular storage depot for rapid lipid mobilization in response to increased cellular demands. LDs have a unique ultrastructure, consisting of a core of neutral lipids encircled by a phospholipid monolayer that is decorated with integral and peripheral proteins. The LD proteome contains numerous lipid metabolic enzymes, regulatory scaffold proteins, proteins involved in LD clustering and fusion, and other proteins of unknown functions. The cellular role of LDs is inherently determined by the composition of its proteome and alteration of the LD protein coat provides a powerful mechanism to adapt LDs to fluctuating metabolic states. Here, we review the current understanding of the molecular mechanisms that govern LD protein targeting and degradation. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Kirill Bersuker
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA
| | - James A Olzmann
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
43
|
Abstract
Lipid droplets are the universal cellular organelles for the transient or long-term storage of lipids. The number, size and composition of lipid droplets vary greatly within cells in a homogenous population as well as in different cell types. The variability of intracellular lipid-storage organelles reflects the diversification of lipid droplet composition and function. Lipid droplet diversification results, for example, in two cellular lipid droplet populations that are prone to diminish and grow, respectively. The aberrant accumulation or depletion of lipids are hallmarks or causes of various human pathologies. Thus, a better understanding of the origins of lipid droplet diversification is not only a fascinating cell biology question but also potentially serves to improve comprehension of pathologies that entail the accumulation of lipids. This Commentary covers the lipid droplet life cycle and highlights the early steps during lipid droplet biogenesis, which we propose to be the potential driving forces of lipid droplet diversification.
Collapse
Affiliation(s)
- Abdou Rachid Thiam
- Laboratoire de Physique Statistique, École Normale Supérieure, PSL Research University; Université Paris Diderot Sorbonne Paris-Cité; Sorbonne Universités UPMC Univ Paris 06; CNRS; 24 rue Lhomond, Paris 75005, France
| | - Mathias Beller
- Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University, Universitätsstr. 1, Düsseldorf 40225, Germany .,Systems Biology of Lipid Metabolism, Heinrich Heine University, Universitätsstr. 1, Düsseldorf 40225, Germany
| |
Collapse
|
44
|
Thul PJ, Tschapalda K, Kolkhof P, Thiam AR, Oberer M, Beller M. Lipid droplet subset targeting of the Drosophila protein CG2254/dmLdsdh1. J Cell Sci 2017; 130:3141-3157. [DOI: 10.1242/jcs.199661] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 07/26/2017] [Indexed: 01/02/2023] Open
Abstract
Lipid droplets (LDs) are the principal organelles of lipid storage. They consist of a hydrophobic core of storage lipids, surrounded by a phospholipid monolayer with proteins attached. While some of these proteins are essential to regulate cellular and organismic lipid metabolism, key questions concerning LD protein function, such as their targeting to LDs, are still unanswered. Intriguingly, some proteins are restricted to LD subsets by an as yet unknown mechanism. This finding makes LD targeting even more complex.
Here, we characterize the Drosophila protein CG2254 which targets LD subsets in cultured cells and different larval Drosophila tissues, where the prevalence of LD subsets appears highly dynamic. We find that an amphipathic amino acid stretch mediates CG2254 LD localization. Additionally, we identified a juxtaposed sequence stretch limiting CG2254 localization to LD subsets. This sequence is sufficient to restrict a chimeric protein - consisting of the subset targeting sequence introduced to an otherwise pan LD localized protein sequence - to LD subsets. Based on its subcellular localization and annotated function, we suggest to rename CG2254 to Lipid droplet subset dehydrogenase 1 (Ldsdh1).
Collapse
Affiliation(s)
- Peter J. Thul
- Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Molecular Developmental Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Kirsten Tschapalda
- Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Molecular Developmental Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Department of Chemical Biology, Max Planck Institute for Molecular Physiology, Dortmund, Germany
| | - Petra Kolkhof
- Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Abdou Rachid Thiam
- Laboratoire de Physique Statistique, Ecole Normale Superieure, PSL Research University, Universite de Paris Diderot Sorbonne Paris-Cite, Paris, France
| | - Monika Oberer
- Institute of Molecular Biosciences, BioTechMed Graz, University of Graz, Austria
| | - Mathias Beller
- Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
45
|
The Lipid Droplet and the Endoplasmic Reticulum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 997:111-120. [DOI: 10.1007/978-981-10-4567-7_8] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
46
|
To M, Peterson CWH, Roberts MA, Counihan JL, Wu TT, Forster MS, Nomura DK, Olzmann JA. Lipid disequilibrium disrupts ER proteostasis by impairing ERAD substrate glycan trimming and dislocation. Mol Biol Cell 2016; 28:270-284. [PMID: 27881664 PMCID: PMC5231896 DOI: 10.1091/mbc.e16-07-0483] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 11/09/2016] [Accepted: 11/15/2016] [Indexed: 12/12/2022] Open
Abstract
The endoplasmic reticulum (ER) mediates the folding, maturation, and deployment of the secretory proteome. Proteins that fail to achieve their native conformation are retained in the ER and targeted for clearance by ER-associated degradation (ERAD), a sophisticated process that mediates the ubiquitin-dependent delivery of substrates to the 26S proteasome for proteolysis. Recent findings indicate that inhibition of long-chain acyl-CoA synthetases with triacsin C, a fatty acid analogue, impairs lipid droplet (LD) biogenesis and ERAD, suggesting a role for LDs in ERAD. However, whether LDs are involved in the ERAD process remains an outstanding question. Using chemical and genetic approaches to disrupt diacylglycerol acyltransferase (DGAT)-dependent LD biogenesis, we provide evidence that LDs are dispensable for ERAD in mammalian cells. Instead, our results suggest that triacsin C causes global alterations in the cellular lipid landscape that disrupt ER proteostasis by interfering with the glycan trimming and dislocation steps of ERAD. Prolonged triacsin C treatment activates both the IRE1 and PERK branches of the unfolded protein response and ultimately leads to IRE1-dependent cell death. These findings identify an intimate relationship between fatty acid metabolism and ER proteostasis that influences cell viability.
Collapse
Affiliation(s)
- Milton To
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720
| | - Clark W H Peterson
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720
| | - Melissa A Roberts
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720
| | - Jessica L Counihan
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720.,Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Tiffany T Wu
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720
| | - Mercedes S Forster
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720
| | - Daniel K Nomura
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720.,Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720.,Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - James A Olzmann
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
47
|
Mishra S, Khaddaj R, Cottier S, Stradalova V, Jacob C, Schneiter R. Mature lipid droplets are accessible to ER luminal proteins. J Cell Sci 2016; 129:3803-3815. [PMID: 27591256 DOI: 10.1242/jcs.189191] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 08/17/2016] [Indexed: 01/14/2023] Open
Abstract
Lipid droplets are found in most organisms where they serve to store energy in the form of neutral lipids. They are formed at the endoplasmic reticulum (ER) membrane where the neutral-lipid-synthesizing enzymes are located. Recent results indicate that lipid droplets remain functionally connected to the ER membrane in yeast and mammalian cells to allow the exchange of both lipids and integral membrane proteins between the two compartments. The precise nature of the interface between the ER membrane and lipid droplets, however, is still ill-defined. Here, we probe the topology of lipid droplet biogenesis by artificially targeting proteins that have high affinity for lipid droplets to inside the luminal compartment of the ER. Unexpectedly, these proteins still localize to lipid droplets in both yeast and mammalian cells, indicating that lipid droplets are accessible from within the ER lumen. These data are consistent with a model in which lipid droplets form a specialized domain in the ER membrane that is accessible from both the cytosolic and the ER luminal side.
Collapse
Affiliation(s)
- Shirish Mishra
- University of Fribourg, Department of Biology, Fribourg 1700, Switzerland
| | - Rasha Khaddaj
- University of Fribourg, Department of Biology, Fribourg 1700, Switzerland
| | - Stéphanie Cottier
- University of Fribourg, Department of Biology, Fribourg 1700, Switzerland
| | - Vendula Stradalova
- University of Fribourg, Department of Biology, Fribourg 1700, Switzerland
| | - Claire Jacob
- University of Fribourg, Department of Biology, Fribourg 1700, Switzerland
| | - Roger Schneiter
- University of Fribourg, Department of Biology, Fribourg 1700, Switzerland
| |
Collapse
|
48
|
Zhang S, Wang Y, Cui L, Deng Y, Xu S, Yu J, Cichello S, Serrero G, Ying Y, Liu P. Morphologically and Functionally Distinct Lipid Droplet Subpopulations. Sci Rep 2016; 6:29539. [PMID: 27386790 PMCID: PMC4937419 DOI: 10.1038/srep29539] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 06/20/2016] [Indexed: 12/12/2022] Open
Abstract
Lipid droplet (LD), a multi-functional organelle, is often found to associate with other cellular membranous structures and vary in size in a given cell, which may be related to their functional diversity. Here we established a method to separate LD subpopulations from isolated CHO K2 LDs into three different size categories. The subpopulation with smallest LDs was nearly free of ER and other membranous structures while those with larger LDs contained intact ER. These distinct subpopulations of LDs differed in their protein composition and ability to recruit proteins. This method was also applicable to LDs obtained from other sources, such as Huh7 cells, mouse liver and brown adipose tissue, et al. We developed an in vitro assay requiring only isolated LDs, Coenzyme A, and ATP to drive lipid synthesis. The LD subpopulation nearly depleted of ER was able to incorporate fatty acids into triacylglycerol and phospholipids. Together, our data demonstrate that LDs in a given cell are heterogeneous in size and function, and suggest that LDs are one of cellular lipid synthetic organelles.
Collapse
Affiliation(s)
- Shuyan Zhang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yang Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Liujuan Cui
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Yaqin Deng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinhai Yu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Simon Cichello
- School of Life Sciences, La Trobe University, Melbourne Victoria 3086, Australia
| | | | - Yunshu Ying
- University of Texas Southwestern Medical Center, Dallas 75390, USA
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
49
|
Peroxin-dependent targeting of a lipid-droplet-destined membrane protein to ER subdomains. Nat Cell Biol 2016; 18:740-51. [PMID: 27295553 PMCID: PMC4925261 DOI: 10.1038/ncb3373] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 05/11/2016] [Indexed: 12/23/2022]
Abstract
Lipid droplets (LDs) are endoplasmic reticulum (ER)-derived lipid storage organelles uniquely encapsulated by phospholipid monolayers. LD membrane proteins are embedded into the monolayer in a monotopic hairpin-topology and therefore likely have requirements for their biogenesis distinct from those inserting as bitopic and polytopic proteins into phospholipid bilayers. UBXD8 belongs to a subfamily of hairpin-proteins that localize to both the ER and LDs, and are initially inserted into the cytoplasmic leaflet of the ER bilayer before partitioning to the LD monolayer. The molecular machinery responsible for inserting hairpin-proteins into membranes, however, is unknown. Here, we report that newly synthesized UBXD8 is posttranslationally inserted into discrete ER-subdomains by a mechanism requiring cytosolic PEX19 and membrane-integrated PEX3, proteins hitherto exclusively implicated in peroxisome biogenesis. Farnesylation of PEX19 uncouples ER/LD- and peroxisome targeting, expanding the function of this peroxin to an ER targeting pathway and suggesting a coordinated biogenesis of LDs and peroxisomes.
Collapse
|
50
|
Abstract
The endoplasmic reticulum is the port of entry for proteins into the secretory pathway and the site of synthesis for several important lipids, including cholesterol, triacylglycerol, and phospholipids. Protein production within the endoplasmic reticulum is tightly regulated by a cohort of resident machinery that coordinates the folding, modification, and deployment of secreted and integral membrane proteins. Proteins failing to attain their native conformation are degraded through the endoplasmic reticulum-associated degradation (ERAD) pathway via a series of tightly coupled steps: substrate recognition, dislocation, and ubiquitin-dependent proteasomal destruction. The same ERAD machinery also controls the flux through various metabolic pathways by coupling the turnover of metabolic enzymes to the levels of key metabolites. We review the current understanding and biological significance of ERAD-mediated regulation of lipid metabolism in mammalian cells.
Collapse
Affiliation(s)
- Julian Stevenson
- Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720;
| | - Edmond Y Huang
- Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720;
| | - James A Olzmann
- Program in Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720;
| |
Collapse
|