1
|
Heikelä H, Mairinoja L, Ruohonen ST, Rytkönen KT, de Brot S, Laiho A, Koskinen S, Suomi T, Elo LL, Strauss L, Poutanen M. Disruption of HSD17B12 in mouse hepatocytes leads to reduced body weight and defect in the lipid droplet expansion associated with microvesicular steatosis. FASEB J 2024; 38:e70034. [PMID: 39248019 DOI: 10.1096/fj.202400333rr] [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: 02/13/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
The function of hydroxysteroid dehydrogenase 12 (HSD17B12) in lipid metabolism is poorly understood. To study this further, we created mice with hepatocyte-specific knockout of HSD17B12 (LiB12cKO). From 2 months on, these mice showed significant fat accumulation in their liver. As they aged, they also had a reduced whole-body fat percentage. Interestingly, the liver fat accumulation did not result in the typical formation of large lipid droplets (LD); instead, small droplets were more prevalent. Thus, LiB12KO liver did not show increased macrovesicular steatosis with the increasing fat content, while microvesicular steatosis was the predominant feature in the liver. This indicates a failure in the LD expansion. This was associated with liver damage, presumably due to lipotoxicity. Notably, the lipidomics data did not support an essential role of HSD17B12 in fatty acid (FA) elongation. However, we did observe a decrease in the quantity of specific lipid species that contain FAs with carbon chain lengths of 18 and 20 atoms, including oleic acid. Of these, phosphatidylcholine and phosphatidylethanolamine have been shown to play a key role in LD formation, and a limited amount of these lipids could be part of the mechanism leading to the dysfunction in LD expansion. The increase in the Cidec expression further supported the deficiency in LD expansion in the LiB12cKO liver. This protein is crucial for the fusion and growth of LDs, along with the downregulation of several members of the major urinary protein family of proteins, which have recently been shown to be altered during endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Hanna Heikelä
- Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Mairinoja
- Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Suvi T Ruohonen
- Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Kalle T Rytkönen
- Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Simone de Brot
- COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Asta Laiho
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Satu Koskinen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Tomi Suomi
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Laura L Elo
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, Finland
| | - Leena Strauss
- Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Matti Poutanen
- Research Centre for Integrative Physiology and Pharmacology and Turku Center for Disease Modeling, Institute of Biomedicine, University of Turku, Turku, Finland
- Department of Internal Medicine and Clinical Nutrition, Centre for Bone and Arthritis Research, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
2
|
Zhang Z, Yu Z, Liang D, Song K, Kong X, He M, Liao X, Huang Z, Kang A, Bai R, Ren Y. Roles of lipid droplets and related proteins in metabolic diseases. Lipids Health Dis 2024; 23:218. [PMID: 39030618 PMCID: PMC11264848 DOI: 10.1186/s12944-024-02212-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Lipid droplets (LDs), which are active organelles, derive from the monolayer membrane of the endoplasmic reticulum and encapsulate neutral lipids internally. LD-associated proteins like RAB, those in the PLIN family, and those in the CIDE family participate in LD formation and development, and they are active players in various diseases, organelles, and metabolic processes (i.e., obesity, non-alcoholic fatty liver disease, and autophagy). Our synthesis on existing research includes insights from the formation of LDs to their mechanisms of action, to provide an overview needed for advancing research into metabolic diseases and lipid metabolism.
Collapse
Affiliation(s)
- Zhongyang Zhang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Zhenghang Yu
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Dianyuan Liang
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Ke Song
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Xiangxin Kong
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Ming He
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China
| | - Xinxin Liao
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Ziyan Huang
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Aijia Kang
- Institute of Hepatobiliary Pancreatic Intestinal Diseases, North Sichuan Medical College, Nanchong, 637000, China
| | - Rubing Bai
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China.
| | - Yixing Ren
- Department of Gastroenterology, Affiliated Hospital of North Sichuan Medical College, South Maoyuan Road, Shunqing District, Nanchong, Sichuan Province, 637000, China.
- General Surgery, Chengdu XinHua Hospital Affiliated to North Sichuan Medical College, Chengdu, 610000, China.
| |
Collapse
|
3
|
Safi R, Menéndez P, Pol A. Lipid droplets provide metabolic flexibility for cancer progression. FEBS Lett 2024; 598:1301-1327. [PMID: 38325881 DOI: 10.1002/1873-3468.14820] [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: 09/04/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024]
Abstract
A hallmark of cancer cells is their remarkable ability to efficiently adapt to favorable and hostile environments. Due to a unique metabolic flexibility, tumor cells can grow even in the absence of extracellular nutrients or in stressful scenarios. To achieve this, cancer cells need large amounts of lipids to build membranes, synthesize lipid-derived molecules, and generate metabolic energy in the absence of other nutrients. Tumor cells potentiate strategies to obtain lipids from other cells, metabolic pathways to synthesize new lipids, and mechanisms for efficient storage, mobilization, and utilization of these lipids. Lipid droplets (LDs) are the organelles that collect and supply lipids in eukaryotes and it is increasingly recognized that the accumulation of LDs is a new hallmark of cancer cells. Furthermore, an active role of LD proteins in processes underlying tumorigenesis has been proposed. Here, by focusing on three major classes of LD-resident proteins (perilipins, lipases, and acyl-CoA synthetases), we provide an overview of the contribution of LDs to cancer progression and discuss the role of LD proteins during the proliferation, invasion, metastasis, apoptosis, and stemness of cancer cells.
Collapse
Affiliation(s)
- Rémi Safi
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Spain
| |
Collapse
|
4
|
Fu X, Zhang S, Liu P. Co-immunoprecipitation for identifying protein-protein interaction on lipid droplets. BIOPHYSICS REPORTS 2024; 10:102-110. [PMID: 38774355 PMCID: PMC11103721 DOI: 10.52601/bpr.2024.240007] [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: 01/27/2024] [Accepted: 02/23/2024] [Indexed: 05/24/2024] Open
Abstract
The lipid droplet (LD) is a conserved organelle that exists in almost all organisms, ranging from bacteria to mammals. Dysfunctions in LDs are linked to a range of human metabolic syndromes. The formation of protein complexes on LDs is crucial for maintaining their function. Investigating how proteins interact on LDs is essential for understanding the role of LDs. We have developed an effective method to uncover protein-protein interactions and protein complexes specifically on LDs. In this method, we conduct co-immunoprecipitation (co-IP) experiments using LD proteins extracted directly from isolated LDs, rather than utilizing proteins from cell lysates. To elaborate, we begin by purifying LDs with high-quality and extracting LD-associated proteins. Subsequently, the co-IP experiment is performed on these LD-associated proteins directly, which would enhance the co-IP experiment specificity of LD-associated proteins. This method enables researchers to directly unveil protein complexes on LDs and gain deeper insights into the functional roles of proteins associated with LDs.
Collapse
Affiliation(s)
- Xiaochuan Fu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyan Zhang
- Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Beijing Institute of Infectious Diseases, Beijing 100015, China
| | - Pingsheng Liu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
5
|
Cabodevilla AG, Son N, Goldberg IJ. Intracellular lipase and regulation of the lipid droplet. Curr Opin Lipidol 2024; 35:85-92. [PMID: 38447014 PMCID: PMC10919935 DOI: 10.1097/mol.0000000000000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
PURPOSE OF REVIEW Lipid droplets are increasingly recognized as distinct intracellular organelles that have functions exclusive to the storage of energetic lipids. Lipid droplets modulate macrophage inflammatory phenotype, control the availability of energy for muscle function, store excess lipid, sequester toxic lipids, modulate mitochondrial activity, and allow transfer of fatty acids between tissues. RECENT FINDINGS There have been several major advances in our understanding of the formation, dissolution, and function of this organelle during the past two years. These include new information on movement and partition of amphipathic proteins between the cytosol and lipid droplet surface, molecular determinants of lipid droplet formation, and pathways leading to lipid droplet hydrophobic lipid formation. Rapid advances in mitochondrial biology have also begun to define differences in their function and partnering with lipid droplets to modulate lipid storage versus oxidation. SUMMARY This relationship of lipid droplets biology and cellular function provides new understanding of an important cellular organelle that influences muscle function, adipose lipid storage, and diseases of lipotoxicity.
Collapse
Affiliation(s)
- Ainara G Cabodevilla
- Division of Endocrinology, New York University Grossman School of Medicine, New York, New York, USA
| | | | | |
Collapse
|
6
|
Yazıcı D, Demir SÇ, Sezer H. Insulin Resistance, Obesity, and Lipotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:391-430. [PMID: 39287860 DOI: 10.1007/978-3-031-63657-8_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Lipotoxicity, originally used to describe the destructive effects of excess fat accumulation on glucose metabolism, causes functional impairments in several metabolic pathways, both in adipose tissue and peripheral organs, like liver, heart, pancreas, and muscle. Ectopic lipid accumulation in the kidneys, liver, and heart has important clinical counterparts like diabetic nephropathy in type 2 diabetes mellitus, obesity-related glomerulopathy, nonalcoholic fatty liver disease, and cardiomyopathy. Insulin resistance due to lipotoxicity indirectly lead to reproductive system disorders, like polycystic ovary syndrome. Lipotoxicity has roles in insulin resistance and pancreatic beta-cell dysfunction. Increased circulating levels of lipids and the metabolic alterations in fatty acid utilization and intracellular signaling have been related to insulin resistance in muscle and liver. Different pathways, like novel protein kinase c pathways and the JNK-1 pathway, are involved as the mechanisms of how lipotoxicity leads to insulin resistance in nonadipose tissue organs, such as liver and muscle. Mitochondrial dysfunction plays a role in the pathogenesis of insulin resistance. Endoplasmic reticulum stress, through mainly increased oxidative stress, also plays an important role in the etiology of insulin resistance, especially seen in non-alcoholic fatty liver disease. Visceral adiposity and insulin resistance both increase the cardiometabolic risk, and lipotoxicity seems to play a crucial role in the pathophysiology of these associations.
Collapse
Affiliation(s)
- Dilek Yazıcı
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey.
| | - Selin Çakmak Demir
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey
| | - Havva Sezer
- Koç University Medical School, Section of Endocrinology and Metabolism, Koç University Hospital, Topkapi, Istanbul, Turkey
| |
Collapse
|
7
|
Engin A. Nonalcoholic Fatty Liver Disease and Staging of Hepatic Fibrosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:539-574. [PMID: 39287864 DOI: 10.1007/978-3-031-63657-8_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is in parallel with the obesity epidemic, and it is the most common cause of liver diseases. The patients with severe insulin-resistant diabetes having high body mass index (BMI), high-grade adipose tissue insulin resistance, and high hepatocellular triacylglycerols (triglycerides; TAG) content develop hepatic fibrosis within a 5-year follow-up. Insulin resistance with the deficiency of insulin receptor substrate-2 (IRS-2)-associated phosphatidylinositol 3-kinase (PI3K) activity causes an increase in intracellular fatty acid-derived metabolites such as diacylglycerol (DAG), fatty acyl CoA, or ceramides. Lipotoxicity-related mechanism of NAFLD could be explained still best by the "double-hit" hypothesis. Insulin resistance is the major mechanism in the development and progression of NAFLD/nonalcoholic steatohepatitis (NASH). Metabolic oxidative stress, autophagy, and inflammation induce NASH progression. In the "first hit" the hepatic concentrations of diacylglycerol increase with an increase in saturated liver fat content in human NAFLD. Activities of mitochondrial respiratory chain complexes are decreased in the liver tissue of patients with NASH. Hepatocyte lipoapoptosis is a critical feature of NASH. In the "second hit," reduced glutathione levels due to oxidative stress lead to the overactivation of c-Jun N-terminal kinase (JNK)/c-Jun signaling that induces cell death in the steatotic liver. Accumulation of toxic levels of reactive oxygen species (ROS) is caused at least by two ineffectual cyclical pathways. First is the endoplasmic reticulum (ER) oxidoreductin (Ero1)-protein disulfide isomerase oxidation cycle through the downstream of the inner membrane mitochondrial oxidative metabolism and the second is the Kelch like-ECH-associated protein 1 (Keap1)-nuclear factor (erythroid-derived 2)-like 2 (Nrf2) pathways. In clinical practice, on ultrasonographic examination, the elevation of transaminases, γ-glutamyltransferase, and the aspartate transaminase to platelet ratio index indicates NAFLD. Fibrosis-4 index, NAFLD fibrosis score, and cytokeratin18 are used for grading steatosis, staging fibrosis, and discriminating the NASH from simple steatosis, respectively. In addition to ultrasonography, "controlled attenuation parameter," "magnetic resonance imaging proton-density fat fraction," "ultrasound-based elastography," "magnetic resonance elastography," "acoustic radiation force impulse elastography imaging," "two-dimensional shear-wave elastography with supersonic imagine," and "vibration-controlled transient elastography" are recommended as combined tests with serum markers in the clinical evaluation of NAFLD. However, to confirm the diagnosis of NAFLD, a liver biopsy is the gold standard. Insulin resistance-associated hyperinsulinemia directly accelerates fibrogenesis during NAFLD development. Although hepatocyte lipoapoptosis is a key driving force of fibrosis progression, hepatic stellate cells and extracellular matrix cells are major fibrogenic effectors. Thereby, these are pharmacological targets of therapies in developing hepatic fibrosis. Nonpharmacological management of NAFLD mainly consists of two alternatives: lifestyle modification and metabolic surgery. Many pharmacological agents that are thought to be effective in the treatment of NAFLD have been tried, but due to lack of ability to attenuate NAFLD, or adverse effects during the phase trials, the vast majority could not be licensed.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
8
|
Pan J, Jin Y, Jin H, Li C, Zhang Y, Liu Y, Jin G, Zhao J, He L, Sheng L. New insights into the function of lipid droplet-related proteins and lipid metabolism of salt-stimulated porcine biceps femoris: label-free quantitative phosphoproteomics, morphometry and bioinformatics. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:7517-7528. [PMID: 37440710 DOI: 10.1002/jsfa.12857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/22/2023] [Accepted: 07/14/2023] [Indexed: 07/15/2023]
Abstract
BACKGROUND Lipid droplets (LDs) are important multifunctional organelles responsible for lipid metabolism of postmortem muscle. However, the dynamics in their building blocks (cores and layers) and phosphorylation of lipid droplet-related proteins (LDRPs) regulating meat lipolysis remain unknown at salt-stimulated conditions. RESULTS LDRPs extracted from cured porcine biceps femoris (1% and 3% salt) were subjected to label-free quantitative phosphoproteomic analysis and LDs morphological validation. Results indicated that 3% salt curing significantly decreased triglyceride (TG) content with increase in glycerol and decrease in LDs fluorescence compared to 1% salt curing. Comparative phosphoproteomics showed that there were significant changes in phosphorylation at 386 sites on 174 LDRPs between assayed groups (P < 0.05). These differential proteins were mainly involved in lipid and carbohydrate metabolism. Curing of 3% salt induced more site-specific phosphorylation of perilipin 1 (PLIN1, at Ser81) and adipose triglyceride lipase (ATGL, at Ser399) than 1%, whereas the phosphorylation (at Ser600) of hormone-sensitive lipase (HSL) was up-regulated. Ultrastructure imaging showed that LDs were mostly associated with mitochondria, and the average diameter of LDs decreased from 2.34 μm (1% salt) to 1.73 μm (3% salt). CONCLUSION Phosphoproteomics unraveled salt-stimulated LDRPs phosphorylation of cured porcine meat provoked intensified lipolysis. Curing of 3% salt allowed an enhanced lipolysis than 1% by up-regulating the phosphorylation sites of LDRPs and recruited lipases. The visible splitting of LDs, together with sarcoplasmic disorganization, supported the lipolysis robustness following 3% salt curing. The finding provides optimization ideas for high-quality production of cured meat products. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiajing Pan
- School of Food and Health, Beijing Technology and Business University, Beijing, China
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yongguo Jin
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Haobo Jin
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Chengliang Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Yan Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Yuanyi Liu
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Guofeng Jin
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Jianying Zhao
- College of Tea and Food Science Technology, Jiangsu Vocational College Agriculture and Forestry, Jurong, China
| | - Lichao He
- School of Food and Health, Beijing Technology and Business University, Beijing, China
| | - Long Sheng
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
9
|
Yang Q, Loureiro ZY, Desai A, DeSouza T, Li K, Wang H, Nicoloro SM, Solivan-Rivera J, Corvera S. Regulation of lipolysis by 14-3-3 proteins on human adipocyte lipid droplets. PNAS NEXUS 2023; 2:pgad420. [PMID: 38130664 PMCID: PMC10733194 DOI: 10.1093/pnasnexus/pgad420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023]
Abstract
Adipocyte lipid droplets (LDs) play a crucial role in systemic lipid metabolism by storing and releasing lipids to meet the organism's energy needs. Hormonal signals such as catecholamines and insulin act on adipocyte LDs, and impaired responsiveness to these signals can lead to uncontrolled lipolysis, lipotoxicity, and metabolic disease. To investigate the mechanisms that control LD function in human adipocytes, we applied proximity labeling mediated by enhanced ascorbate peroxidase (APEX2) to identify the interactome of PLIN1 in adipocytes differentiated from human mesenchymal progenitor cells. We identified 70 proteins that interact specifically with PLIN1, including PNPLA2 and LIPE, which are the primary effectors of regulated triglyceride hydrolysis, and 4 members of the 14-3-3 protein family (YWHAB, YWHAE, YWHAZ, and YWHAG), which are known to regulate diverse signaling pathways. Functional studies showed that YWHAB is required for maximum cyclic adenosine monophosphate (cAMP)-stimulated lipolysis, as its CRISPR-Cas9-mediated knockout mitigates lipolysis through a mechanism independent of insulin signaling. These findings reveal a new regulatory mechanism operating in human adipocytes that can impact lipolysis and potentially systemic metabolism.
Collapse
Affiliation(s)
- Qin Yang
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester MA 01605, USA
| | - Zinger Yang Loureiro
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Morningside Graduate School of Biomedical Sciences, University of Massachusetts Chan Medical School, Worcester MA 01605, USA
| | - Anand Desai
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Tiffany DeSouza
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Kaida Li
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Hui Wang
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Sarah M Nicoloro
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Javier Solivan-Rivera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Silvia Corvera
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
- Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
10
|
Li J, Wang Y, Yang P, Han H, Zhang G, Xu H, Quan K. Overexpression of ATGL impairs lipid droplet accumulation by accelerating lipolysis in goat mammary epithelial cells. Anim Biotechnol 2023; 34:3126-3134. [PMID: 36306180 DOI: 10.1080/10495398.2022.2136678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Adipose triglyceride lipase (ATGL) is the key enzyme for the degradation of triacylglycerols (TAGs). It functions in concert with other enzymes to mobilize TAG and supply fatty acids (FAs) for energy production. Dysregulated lipolysis leads to excess concentrations of circulating FAs, which may lead to destructive and lipotoxic effects to the organism. To understand the role of ATGL in mammary lipid metabolism, ATGL was overexpressed in goat mammary epithelial cells (GMECs) by using a recombinant adenovirus system. ATGL overexpression decreased lipid droplet (LD) accumulation and cellular TG content (p < 0.05) along with a decrease in the expression of the key enzyme that catalyzes the final step of TG synthesis (DGAT). Significant increases were observed in the expression of genes related to lipolysis (hormone-sensitive lipase [HSL]) and FA desaturation (SCD) by ATGL overexpression. Genes responsible for FA oxidation (PPARα), LD formation and secretion (ADRP and BTN1A1), and long-chain FA uptake (CD36) were all decreased by ATGL overexpression (p < 0.05). The primary products of TAG lipolysis, free FAs (FFAs), were notably increased in the ATGL-overexpressing cells. Taken together, our results demonstrated that ATGL activation impairs lipid formation partially through accelerating lipolysis in GMECs.
Collapse
Affiliation(s)
- Jun Li
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Yaling Wang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, PR China
| | - Pengkun Yang
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Haoyuan Han
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Guizhi Zhang
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| | - Huifen Xu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, PR China
| | - Kai Quan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou, PR China
| |
Collapse
|
11
|
Schiffmann A, Ahlswede L, Gimpl G. Reversible translocation of acyl-CoA:cholesterol acyltransferase (ACAT) between the endoplasmic reticulum and vesicular structures. Front Mol Biosci 2023; 10:1258799. [PMID: 38028547 PMCID: PMC10667705 DOI: 10.3389/fmolb.2023.1258799] [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: 07/14/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
The enzyme acyl-CoA:cholesterol acyltransferase (ACAT) is normally localized in the endoplasmic reticulum (ER) where it can esterify cholesterol for storage in lipid droplets and/or the formation of lipoproteins. Here, we report that ACAT can translocate from the ER into vesicular structures in response to different ACAT inhibitors. The translocation was fast (within minutes), reversible and occurred in different cell types. Interestingly, oleic acid was able to fasten the re-translocation from vesicles back into the reticular ER network. The process of ACAT translocation could also be induced by cyclodextrins, cholesterol, lanosterol (but not 4-cholestene-3 one), 25-hydroxycholesterol, and by certain stress stimuli such as hyperosmolarity (sucrose treatment), temperature change, or high-density cultivation. In vitro esterification showed that ACAT remains fully active after it has been translocated to vesicles in response to hyperosmotic sucrose treatment of the cells. The translocation process was not accompanied by changes in the electrophoretic mobility of ACAT, even after chemical crosslinking. Interestingly, the protein synthesis inhibitor cycloheximide showed a stimulating effect on ACAT activity and prevented the translocation of ACAT from the ER into vesicles.
Collapse
Affiliation(s)
| | | | - Gerald Gimpl
- Department of Chemistry and Biochemistry, Biocenter II, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
12
|
Renier TJ, Paetz OR, Paal MC, Long AB, Brown MR, Vuong SH, Perumal SK, Kharbanda KK, Listenberger LL. Changing the phospholipid composition of lipid droplets alters localization of select lipid droplet proteins. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000960. [PMID: 38021172 PMCID: PMC10656625 DOI: 10.17912/micropub.biology.000960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/30/2023] [Accepted: 10/31/2023] [Indexed: 12/01/2023]
Abstract
Our experiments aim to determine if decreasing the amount of phosphatidylcholine (PC) relative to phosphatidylethanolamine (PE) at the lipid droplet surface changes the localization of specific lipid droplet proteins. We manipulate lipid droplet phospholipids in both a cultured mouse hepatocyte (AML12) cell line and on synthetic lipid droplets. Decreasing the PC:PE ratio increases perilipin 2, decreases DGAT2, and does not change rab18 or lanosterol synthase levels on lipid droplets. These differences may be explained by the distinct structural motifs that mediate the protein-lipid droplet interactions.
Collapse
Affiliation(s)
- Timothy J. Renier
- Departments of Biology and Chemistry, St. Olaf College, Northfield, Minnesota, United States
| | - Olivia R. Paetz
- Departments of Biology and Chemistry, St. Olaf College, Northfield, Minnesota, United States
| | - Matthew C. Paal
- Departments of Biology and Chemistry, St. Olaf College, Northfield, Minnesota, United States
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Alex B. Long
- Departments of Biology and Chemistry, St. Olaf College, Northfield, Minnesota, United States
| | - Margaret R. Brown
- Departments of Biology and Chemistry, St. Olaf College, Northfield, Minnesota, United States
| | - Sunny H. Vuong
- Departments of Biology and Chemistry, St. Olaf College, Northfield, Minnesota, United States
| | - Sathish Kumar Perumal
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
| | - Kusum K. Kharbanda
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, Nebraska, United States
- Research Service, Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, Nebraska, United States
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, United States
| | - Laura L. Listenberger
- Departments of Biology and Chemistry, St. Olaf College, Northfield, Minnesota, United States
| |
Collapse
|
13
|
Roberts MA, Deol KK, Mathiowetz AJ, Lange M, Leto DE, Stevenson J, Hashemi SH, Morgens DW, Easter E, Heydari K, Nalls MA, Bassik MC, Kampmann M, Kopito RR, Faghri F, Olzmann JA. Parallel CRISPR-Cas9 screens identify mechanisms of PLIN2 and lipid droplet regulation. Dev Cell 2023; 58:1782-1800.e10. [PMID: 37494933 PMCID: PMC10530302 DOI: 10.1016/j.devcel.2023.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 06/01/2023] [Accepted: 07/03/2023] [Indexed: 07/28/2023]
Abstract
Despite the key roles of perilipin-2 (PLIN2) in governing lipid droplet (LD) metabolism, the mechanisms that regulate PLIN2 levels remain incompletely understood. Here, we leverage a set of genome-edited human PLIN2 reporter cell lines in a series of CRISPR-Cas9 loss-of-function screens, identifying genetic modifiers that influence PLIN2 expression and post-translational stability under different metabolic conditions and in different cell types. These regulators include canonical genes that control lipid metabolism as well as genes involved in ubiquitination, transcription, and mitochondrial function. We further demonstrate a role for the E3 ligase MARCH6 in regulating triacylglycerol biosynthesis, thereby influencing LD abundance and PLIN2 stability. Finally, our CRISPR screens and several published screens provide the foundation for CRISPRlipid (http://crisprlipid.org), an online data commons for lipid-related functional genomics data. Our study identifies mechanisms of PLIN2 and LD regulation and provides an extensive resource for the exploration of LD biology and lipid metabolism.
Collapse
Affiliation(s)
- Melissa A Roberts
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kirandeep K Deol
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alyssa J Mathiowetz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mike Lange
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Dara E Leto
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Julian Stevenson
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sayed Hadi Hashemi
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61820, USA
| | - David W Morgens
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emilee Easter
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kartoosh Heydari
- Cancer Research Laboratory FACS Core Facility, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Mike A Nalls
- Data Tecnica International, LLC, Washington, DC, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael C Bassik
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Martin Kampmann
- Institute for Neurodegenerative Diseases, Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ron R Kopito
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Faraz Faghri
- Data Tecnica International, LLC, Washington, DC, USA; Center for Alzheimer's and Related Dementias, National Institutes of Health, Bethesda, MD 20892, USA; Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
14
|
Zhang D, Zou T, Liu Q, Chen J, Xiao M, Zheng A, Zhang Z, Du F, Dai Y, Xiang S, Wu X, Li M, Chen Y, Zhao Y, Shen J, Chen G, Xiao Z. Transcriptomic characterization revealed that METTL7A inhibits melanoma progression via the p53 signaling pathway and immunomodulatory pathway. PeerJ 2023; 11:e15799. [PMID: 37547717 PMCID: PMC10404031 DOI: 10.7717/peerj.15799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 07/05/2023] [Indexed: 08/08/2023] Open
Abstract
METTL7A is a protein-coding gene expected to be associated with methylation, and its expression disorder is associated with a range of diseases. However, few research have been carried out to explore the relationship between METTL7A and tumor malignant phenotype as well as the involvement potential mechanism. We conducted our research via a combination of silico analysis and molecular biology techniques to investigate the biological function of METTL7A in the progression of cancer. Gene expression and clinical information were extracted from the TCGA database to explore expression variation and prognostic value of METTL7A. In vitro, CCK8, transwell, wound healing and colony formation assays were conducted to explore the biological functions of METT7A in cancer cell. GSEA was performed to explore the signaling pathway involved in METTL7A and validated via western blotting. In conclusion, METTL7A was downregulated in most cancer tissues and its low expression was associated with shorter overall survival. In melanoma, METTL7A downregulation was associated with poorer clinical staging, lower levels of TIL infiltration, higher IC50 levels of chemotherapeutic agents, and poorer immunotherapy outcomes. QPCR results confirm that METTL7A is down-regulated in melanoma cells. Cell function assays showed that METTL7A knockdown promoted proliferation, invasion, migration and clone formation of melanoma cells. Mechanistic studies showed that METTL7A inhibits tumorigenicity through the p53 signaling pathway. Meanwhile, METTL7A is also a potential immune regulatory factor.
Collapse
Affiliation(s)
- Duoli Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
| | - Tao Zou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
| | - Qingsong Liu
- Department of Pathology, The First People’s Hospital of Neijiang, Neijiang, China
| | - Jie Chen
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
| | - Mintao Xiao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
| | - Anfu Zheng
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
| | - Zhuo Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
| | - Fukuan Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yalan Dai
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Shixin Xiang
- Department of Pharmacy, University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Xu Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Mingxing Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yu Chen
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Yueshui Zhao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Jing Shen
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| | - Guiquan Chen
- Chinese Medicine Hospital Affiliated to Southwest Medical University, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Laboratory of Molecular Pharmacology, Luzhou, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou, China
- South Sichuan Institute of Translational Medicine, Luzhou, Sichuan, China
| |
Collapse
|
15
|
Dalen KT, Li Y. Regulation of lipid droplets and cholesterol metabolism in adrenal cortical cells. VITAMINS AND HORMONES 2023; 124:79-136. [PMID: 38408810 DOI: 10.1016/bs.vh.2023.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
The adrenal gland is composed of two distinctly different endocrine moieties. The interior medulla consists of neuroendocrine chromaffin cells that secrete catecholamines like adrenaline and noradrenaline, while the exterior cortex consists of steroidogenic cortical cells that produce steroid hormones, such as mineralocorticoids (aldosterone), glucocorticoids (cortisone and cortisol) and androgens. Synthesis of steroid hormones in cortical cells requires substantial amounts of cholesterol, which is the common precursor for steroidogenesis. Cortical cells may acquire cholesterol from de novo synthesis and uptake from circulating low- and high-density lipoprotein particles (LDL and HDL). As cholesterol is part of the plasma membrane in all mammalian cells and an important regulator of membrane fluidity, cellular levels of free cholesterol are tightly regulated. To ensure a robust supply of cholesterol for steroidogenesis and to avoid cholesterol toxicity, cortical cells store large amounts of cholesterol as cholesteryl esters in intracellular lipid droplets. Cortical steroidogenesis relies on both mobilization of cholesterol from lipid droplets and constant uptake of circulating cholesterol to replenish lipid droplet stores. This chapter will describe mechanisms involved in cholesterol uptake, cholesteryl ester synthesis, lipid droplet formation, hydrolysis of stored cholesteryl esters, as well as their impact on steroidogenesis. Additionally, animal models and human diseases characterized by altered cortical cholesteryl ester storage, with or without abnormal steroidogenesis, will be discussed.
Collapse
Affiliation(s)
- Knut Tomas Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Norway; The Norwegian Transgenic Center, Institute of Basic Medical Sciences, University of Oslo, Norway.
| | - Yuchuan Li
- Department of Hepato-Pancreato-Biliary Surgery, Institute of Clinical Medicine, University of Oslo, Norway
| |
Collapse
|
16
|
Hammoudeh N, Soukkarieh C, Murphy DJ, Hanano A. Mammalian lipid droplets: structural, pathological, immunological and anti-toxicological roles. Prog Lipid Res 2023; 91:101233. [PMID: 37156444 DOI: 10.1016/j.plipres.2023.101233] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Mammalian lipid droplets (LDs) are specialized cytosolic organelles consisting of a neutral lipid core surrounded by a membrane made up of a phospholipid monolayer and a specific population of proteins that varies according to the location and function of each LD. Over the past decade, there have been significant advances in the understanding of LD biogenesis and functions. LDs are now recognized as dynamic organelles that participate in many aspects of cellular homeostasis plus other vital functions. LD biogenesis is a complex, highly-regulated process with assembly occurring on the endoplasmic reticulum although aspects of the underpinning molecular mechanisms remain elusive. For example, it is unclear how many enzymes participate in the biosynthesis of the neutral lipid components of LDs and how this process is coordinated in response to different metabolic cues to promote or suppress LD formation and turnover. In addition to enzymes involved in the biosynthesis of neutral lipids, various scaffolding proteins play roles in coordinating LD formation. Despite their lack of ultrastructural diversity, LDs in different mammalian cell types are involved in a wide range of biological functions. These include roles in membrane homeostasis, regulation of hypoxia, neoplastic inflammatory responses, cellular oxidative status, lipid peroxidation, and protection against potentially toxic intracellular fatty acids and lipophilic xenobiotics. Herein, the roles of mammalian LDs and their associated proteins are reviewed with a particular focus on their roles in pathological, immunological and anti-toxicological processes.
Collapse
Affiliation(s)
- Nour Hammoudeh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Denis J Murphy
- School of Applied Sciences, University of South Wales, Pontypridd, CF37 1DL, Wales, United Kingdom..
| | - Abdulsamie Hanano
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria (AECS), P.O. Box 6091, Damascus, Syria..
| |
Collapse
|
17
|
Sun Y, Heng J, Liu F, Zhang S, Liu P. Isolation and proteomic study of fish liver lipid droplets. BIOPHYSICS REPORTS 2023; 9:120-133. [PMID: 38028150 PMCID: PMC10648235 DOI: 10.52601/bpr.2023.230004] [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: 02/26/2023] [Accepted: 06/02/2023] [Indexed: 12/01/2023] Open
Abstract
Lipid droplets (LDs) are a neutral lipid storage organelle that is conserved in almost all species. Excessive storage of neutral lipids in LDs is directly associated with many metabolic syndromes. Zebrafish is a better model animal for the study of LD biology due to its transparent embryonic stage compared to other organisms. However, the study of LDs in fish has been difficult due to the lack of specific LD marker proteins and the limitation of purification technology. In this paper, the purification and proteomic analysis of liver LDs of fish including zebrafish and Carassius auratus were performed for the first time. 259 and 267 proteins were identified respectively. Besides most of the identified proteins were reported in previous LD proteomes of mammals, indicating the similarity between mammal and fish LDs. We also identified many unique proteins of liver LDs in fish that are involved in the regulation of LD dynamics. Through morphological and biochemical analysis, we found that the marker protein Plin2 of zebrafish LD was located on LDs in Huh7 cells. These results will facilitate further study of LDs in fish and liver metabolic diseases using fish as a model animal.
Collapse
Affiliation(s)
- Yuwei Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Heng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
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: 7] [Impact Index Per Article: 7.0] [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
|
19
|
Zhou X, Matskova L, Zheng S, Wang X, Wang Y, Xiao X, Mo Y, Wölke M, Li L, Zheng Q, Huang G, Zhang Z, Ernberg I. Mechanisms of Anergic Inflammatory Response in Nasopharyngeal Carcinoma Cells Despite Ubiquitous Constitutive NF-κB Activation. Front Cell Dev Biol 2022; 10:861916. [PMID: 35938161 PMCID: PMC9353648 DOI: 10.3389/fcell.2022.861916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 06/15/2022] [Indexed: 11/29/2022] Open
Abstract
Commensal microbes cross talk with their colonized mucosa. We show that microbes and their cell wall components induce an inflammatory response in cultured human mucosal cells derived from the nonmalignant nasopharyngeal epithelium (NNE) cells in vitro. NNE cells show significant induction of NF-κB with nuclear shuttling and inflammatory gene response when exposed to Gram-positive bacteria (streptococci) or peptidoglycan (PGN), a component of the Gram-positive bacterial cell wall. This response is abrogated in nasopharyngeal carcinoma (NPC)–derived cell lines. The inflammatory response induced by NF-κB signaling was blocked at two levels in the tumor-derived cells. We found that NF-κB was largely trapped in lipid droplets (LDs) in the cytoplasm of the NPC-derived cells, while the increased expression of lysine-specific histone demethylase 1 (LSD1, a repressive nuclear factor) reduces the response mediated by remaining NF-κB at the promoters responding to inflammatory stimuli. This refractory response in NPC cells might be a consequence of long-term exposure to microbes in vivo during carcinogenic progression. It may contribute to the decreased antitumor immune responses in NPC, among others despite heavy T-helper cell infiltration, and thus facilitate tumor progression.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Liudmila Matskova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Shixing Zheng
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Xiaoxia Wang
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Yifang Wang
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xue Xiao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yingxi Mo
- Department of Research, Affiliated Tumor Hospital of Guangxi Medical University, Nanning, China
| | - Marleen Wölke
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Limei Li
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Qian Zheng
- Life Science Institute, Guangxi Medical University, Nanning, China
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Guangwu Huang
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhe Zhang
- Key Laboratory of Early Prevention and Treatment for Regional High-Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
- ENT Institute and Department of Otorhinolaryngology, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Ingemar Ernberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
- *Correspondence: Ingemar Ernberg,
| |
Collapse
|
20
|
Sánchez-Álvarez M, del Pozo MÁ, Bosch M, Pol A. Insights Into the Biogenesis and Emerging Functions of Lipid Droplets From Unbiased Molecular Profiling Approaches. Front Cell Dev Biol 2022; 10:901321. [PMID: 35756995 PMCID: PMC9213792 DOI: 10.3389/fcell.2022.901321] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/17/2022] [Indexed: 11/30/2022] Open
Abstract
Lipid droplets (LDs) are spherical, single sheet phospholipid-bound organelles that store neutral lipids in all eukaryotes and some prokaryotes. Initially conceived as relatively inert depots for energy and lipid precursors, these highly dynamic structures play active roles in homeostatic functions beyond metabolism, such as proteostasis and protein turnover, innate immunity and defense. A major share of the knowledge behind this paradigm shift has been enabled by the use of systematic molecular profiling approaches, capable of revealing and describing these non-intuitive systems-level relationships. Here, we discuss these advances and some of the challenges they entail, and highlight standing questions in the field.
Collapse
Affiliation(s)
- Miguel Sánchez-Álvarez
- Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel del Pozo
- Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Marta Bosch
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
21
|
Liu H, Du J, Chao S, Li S, Cai H, Zhang H, Chen G, Liu P, Bu P. Fusobacterium nucleatum Promotes Colorectal Cancer Cell to Acquire Stem Cell-Like Features by Manipulating Lipid Droplet-Mediated Numb Degradation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2105222. [PMID: 35170250 PMCID: PMC9035998 DOI: 10.1002/advs.202105222] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 12/19/2021] [Indexed: 05/26/2023]
Abstract
Fusobacterium nucleatum is a critical microbe that contributes to colorectal cancer progression and chemoresistance. However, whether and how F. nucleatum regulates colorectal cancer stem-like cells (CCSCs) remains unknown. Here, the authors show that F. nucleatum promotes CCSC self-renewal, and non-CCSCs to acquire CCSC features by manipulating cellular lipid accumulation. F. nucleatum infection decreases lipid accumulation in CCSCs by enhancing fatty acid oxidation, thus promoting CCSC self-renewal. In contrast, F. nucleatum increases lipid accumulation in non-CCSCs by promoting fatty acid formation. Lipids are deposited as lipid droplets, which recruits Numb, a key cell fate regulator, through the AP2A/ACSL3 complex, and MDM2, an E3 ubiquitin ligase, though VCP and UBXD8. On lipid droplets, Numb is degraded by MDM2, activating Notch signaling, thus promoting gain of stem-like cell features. Their findings demonstrate that F. nucleatum directly manipulates colorectal cancer cell fate and reveal the mechanism of lipid droplet-mediated Numb degradation for activating Notch signaling.
Collapse
Affiliation(s)
- Haiyang Liu
- Key Laboratory of RNA BiologyKey Laboratory of Protein and Peptide PharmaceuticalInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
| | - Junfeng Du
- Department of General Surgerythe 7th Medical CenterChinese PLA General HospitalBeijing100700China
- The 2nd School of Clinical MedicineSouthern Medical UniversityGuangdong510515China
- Medical Department of General Surgerythe 1st Medical CenterChinese PLA General HospitalBeijing100853China
| | - Shanshan Chao
- Key Laboratory of RNA BiologyKey Laboratory of Protein and Peptide PharmaceuticalInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijing100049China
| | - Shuoguo Li
- Center for Biological ImagingInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
| | - Huiyun Cai
- Department of General Surgerythe 7th Medical CenterChinese PLA General HospitalBeijing100700China
| | - Hongjie Zhang
- The core facilityInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
| | - Gang Chen
- Department of General Surgerythe 7th Medical CenterChinese PLA General HospitalBeijing100700China
- Medical Department of General Surgerythe 1st Medical CenterChinese PLA General HospitalBeijing100853China
| | - Pingsheng Liu
- National Laboratory of BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
- Center for Excellence in BiomacromoleculesChinese Academy of SciencesBeijing100101China
| | - Pengcheng Bu
- Key Laboratory of RNA BiologyKey Laboratory of Protein and Peptide PharmaceuticalInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijing100049China
- Center for Excellence in BiomacromoleculesChinese Academy of SciencesBeijing100101China
| |
Collapse
|
22
|
Protocol for using artificial lipid droplets to study the binding affinity of lipid droplet-associated proteins. STAR Protoc 2022; 3:101214. [PMID: 35265861 PMCID: PMC8899027 DOI: 10.1016/j.xpro.2022.101214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
23
|
Luo S, Yang M, Zhao H, Han Y, Jiang N, Yang J, Chen W, Li C, Liu Y, Zhao C, Sun L. Caveolin-1 Regulates Cellular Metabolism: A Potential Therapeutic Target in Kidney Disease. Front Pharmacol 2021; 12:768100. [PMID: 34955837 PMCID: PMC8703113 DOI: 10.3389/fphar.2021.768100] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/08/2021] [Indexed: 01/09/2023] Open
Abstract
The kidney is an energy-consuming organ, and cellular metabolism plays an indispensable role in kidney-related diseases. Caveolin-1 (Cav-1), a multifunctional membrane protein, is the main component of caveolae on the plasma membrane. Caveolae are represented by tiny invaginations that are abundant on the plasma membrane and that serve as a platform to regulate cellular endocytosis, stress responses, and signal transduction. However, caveolae have received increasing attention as a metabolic platform that mediates the endocytosis of albumin, cholesterol, and glucose, participates in cellular metabolic reprogramming and is involved in the progression of kidney disease. It is worth noting that caveolae mainly depend on Cav-1 to perform the abovementioned cellular functions. Furthermore, the mechanism by which Cav-1 regulates cellular metabolism and participates in the pathophysiology of kidney diseases has not been completely elucidated. In this review, we introduce the structure and function of Cav-1 and its functions in regulating cellular metabolism, autophagy, and oxidative stress, focusing on the relationship between Cav-1 in cellular metabolism and kidney disease; in addition, Cav-1 that serves as a potential therapeutic target for treatment of kidney disease is also described.
Collapse
Affiliation(s)
- Shilu Luo
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Ming Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Hao Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yachun Han
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Na Jiang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Jinfei Yang
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Wei Chen
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Chenrui Li
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Yan Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Chanyue Zhao
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, China
| |
Collapse
|
24
|
Banerjee S, Bose D, Das S, Chatterjee N, Mishra S, Das Saha K. Leishmania donovani infection induce Extracellular signal-regulated kinase ½ (ERK½) mediated lipid droplet generation in macrophages. Mol Immunol 2021; 141:328-337. [PMID: 34953281 DOI: 10.1016/j.molimm.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 12/05/2021] [Accepted: 12/05/2021] [Indexed: 10/19/2022]
Abstract
Recently unfolded mechanisms showed lipid droplet helps in pathogen survival and paralyzes host immune response. In the present study, we showed the extent of lipid droplet(LD) generation in Leishmania donovani infection, the signaling involved, and their function concerning pathogenicity. RAW 264.7 and J774A.1 cells were used to infect with L. donovani and then flow cytometry and confocal microscopy were used to detect lipid droplet generation and subsequent assays. In this study, we showed that L. donovani AG83 (AG83/MHOM/1983) triggers lipid droplet formation in macrophages in a time-dependent manner. We provide novel insight into the signaling molecules which is responsible for LD accumulation. Interestingly, LPG deficient attenuated Leishmania strain UR6 (UR6/MHOM/1978) failed to fuel LD generation. But inhibition of phagosome maturation drastically stimulates LD accumulation in UR6 infected MΦs. Aspirin treatment in AG83 infected MΦs does not only lower LD load but also favors phagolysosome biogenesis and corrects cytokine balance. Employing strategies to circumvent halt in phagosome maturation using drugs that manipulate lipid droplet generation could be used as a therapeutic tool to resist parasite growth in the early hour of infection.
Collapse
Affiliation(s)
- Somenath Banerjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Dipayan Bose
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Subhadip Das
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Nabanita Chatterjee
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Snehasish Mishra
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India
| | - Krishna Das Saha
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, Kolkata, West Bengal, India.
| |
Collapse
|
25
|
A Decade of Mighty Lipophagy: What We Know and What Facts We Need to Know? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5539161. [PMID: 34777688 PMCID: PMC8589519 DOI: 10.1155/2021/5539161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 12/24/2022]
Abstract
Lipids are integral cellular components that act as substrates for energy provision, signaling molecules, and essential constituents of biological membranes along with a variety of other biological functions. Despite their significance, lipid accumulation may result in lipotoxicity, impair autophagy, and lysosomal function that may lead to certain diseases and metabolic syndromes like obesity and even cell death. Therefore, these lipids are continuously recycled and redistributed by the process of selective autophagy specifically termed as lipophagy. This selective form of autophagy employs lysosomes for the maintenance of cellular lipid homeostasis. In this review, we have reviewed the current literature about how lipid droplets (LDs) are recruited towards lysosomes, cross-talk between a variety of autophagy receptors present on LD surface and lysosomes, and lipid hydrolysis by lysosomal enzymes. In addition to it, we have tried to answer most of the possible questions related to lipophagy regulation at different levels. Moreover, in the last part of this review, we have discussed some of the pathological states due to the accumulation of these LDs and their possible treatments under the light of currently available findings.
Collapse
|
26
|
Wei M, Huang X, Bian C, Sun J, Ji H. ATF6-DGAT pathway is involved in TLR7-induced innate immune response in Ctenopharyngodon idellus kidney cells. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 124:104197. [PMID: 34228994 DOI: 10.1016/j.dci.2021.104197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/01/2021] [Accepted: 07/01/2021] [Indexed: 06/13/2023]
Abstract
DGAT1 and DGAT2 are two acyl-CoA:diacylglycerol O-acyltransferase (DGAT) enzymes that catalyze the final step in triglyceride (TG) synthesis. TGs are the primary constituents of lipid droplets (LDs). Although it has been demonstrated that LDs modulate immune and inflammatory responses in CIK cells, little is known about whether DGAT1 and DGAT2 involve in this process. Firstly, grass carp DGAT2 was isolated and characterized, encoding 361 amino acids, and all DGAT2 proteins in genomic structures are conserved in vertebrates. Then, using TLR7 agonist, we induced LDs accumulation in CIK cells. Only DGAT1b and DGAT2 were upregulated in forming TLR7 agonist induced-LDs. Next, we utilized small-molecule inhibitors of DGAT1 and DGAT2. The results indicated that DGAT1 inactivation attenuated TG content and the relative expressions of IFNα3, NF-κB, IL-1β, and TNFα genes, whereas DGAT2 inhibition decreased TG content and the relative expressions of MyD88, IRF7, IFNα3, NF-κB, IL-1β, and TNFα genes, implying that DGAT1-generated LDs and DGAT2-generated LDs contribute to TLR7-induced immune response via different signaling pathways. Finally, inhibiting ATF6 effectively decreased DGAT-generated LDs accumulation and the expression of TLR7 signaling-related genes induced by TLR7 agonist, implying that ATF6 UPR pathway may mediate the role of DGAT-generated LDs in TLR7 signaling. Overall, we demonstrate that DGAT1 and DGAT2-catalyzed TAG synthesis may generate different LDs to provide distinct signaling platforms for innate TLR7 signaling.
Collapse
Affiliation(s)
- Mingkui Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaocheng Huang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chenchen Bian
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Jian Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
27
|
Pereira-Dutra FS, Bozza PT. Lipid droplets diversity and functions in inflammation and immune response. Expert Rev Proteomics 2021; 18:809-825. [PMID: 34668810 DOI: 10.1080/14789450.2021.1995356] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION Lipid droplets (LDs) are dynamic and evolutionary conserved lipid-enriched organelles composed of a core of neutral lipids surrounded by a monolayer of phospholipids associated with a diverse array of proteins that are cell- and stimulus-regulated. Far beyond being simply a deposit of neutral lipids, accumulating evidence demonstrate that LDs act as spatial and temporal local for lipid and protein compartmentalization and signaling organization. AREAS COVERED This review focuses on the progress in our understanding of LD protein diversity and LD functions in the context of cell signaling and immune responses, highlighting the relationship between LD composition with the multiple roles of this organelle in immunometabolism, inflammation and host-response to infection. EXPERT OPINION LDs are essential platforms for various cellular processes, including metabolic regulation, cell signaling, and immune responses. The functions of LD in infection and inflammatory disease are associated with the dynamic and complexity of their proteome. Our contemporary view place LDs as critical regulators of different inflammatory and infectious diseases and key markers of leukocyte activation.
Collapse
Affiliation(s)
- Filipe S Pereira-Dutra
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
28
|
Montana V, Flint D, Waagepetersen HS, Schousboe A, Parpura V. Two Metabolic Fuels, Glucose and Lactate, Differentially Modulate Exocytotic Glutamate Release from Cultured Astrocytes. Neurochem Res 2021; 46:2551-2579. [PMID: 34057673 PMCID: PMC9015689 DOI: 10.1007/s11064-021-03340-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022]
Abstract
Astrocytes have a prominent role in metabolic homeostasis of the brain and can signal to adjacent neurons by releasing glutamate via a process of regulated exocytosis. Astrocytes synthesize glutamate de novo owing to the pyruvate entry to the citric/tricarboxylic acid cycle via pyruvate carboxylase, an astrocyte specific enzyme. Pyruvate can be sourced from two metabolic fuels, glucose and lactate. Thus, we investigated the role of these energy/carbon sources in exocytotic glutamate release from astrocytes. Purified astrocyte cultures were acutely incubated (1 h) in glucose and/or lactate-containing media. Astrocytes were mechanically stimulated, a procedure known to increase intracellular Ca2+ levels and cause exocytotic glutamate release, the dynamics of which were monitored using single cell fluorescence microscopy. Our data indicate that glucose, either taken-up from the extracellular space or mobilized from the intracellular glycogen storage, sustained glutamate release, while the availability of lactate significantly reduced the release of glutamate from astrocytes. Based on further pharmacological manipulation during imaging along with tandem mass spectrometry (proteomics) analysis, lactate alone, but not in the hybrid fuel, caused metabolic changes consistent with an increased synthesis of fatty acids. Proteomics analysis further unveiled complex changes in protein profiles, which were condition-dependent and generally included changes in levels of cytoskeletal proteins, proteins of secretory organelle/vesicle traffic and recycling at the plasma membrane in aglycemic, lactate or hybrid-fueled astrocytes. These findings support the notion that the availability of energy sources and metabolic milieu play a significant role in gliotransmission.
Collapse
Affiliation(s)
- Vedrana Montana
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| | - Daniel Flint
- Luxumbra Strategic Research, LLC, Arlington, VA, USA
| | - Helle S Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
29
|
Ma X, Zhi Z, Zhang S, Zhou C, Mechler A, Liu P. Validating an artificial organelle: Studies of lipid droplet-specific proteins on adiposome platform. iScience 2021; 24:102834. [PMID: 34368652 PMCID: PMC8326204 DOI: 10.1016/j.isci.2021.102834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/28/2021] [Accepted: 07/08/2021] [Indexed: 10/25/2022] Open
Abstract
New strategies are urgently needed to characterize the functions of the lipid droplet (LD). Here, adiposome, an artificial LD mimetic platform, was validated by comparative in vitro bioassays. Scatchard analysis found that the binding of perilipin 2 (PLIN2) to the adiposome surface was saturable. Phosphatidylinositol (PtdIns) was found to inhibit PLIN2 binding while it did not impede perilipin 3 (PLIN3). Structural analysis combined with mutagenesis revealed that the 73rd glutamic acid of PLIN2 is significant for the effect of PtdIns on the PLIN2 binding. Furthermore, adiposome was also found to be an ideal platform for in situ enzymatic activity measurement of adipose triglyceride lipase (ATGL). The significant serine mutants of ATGL were found to cause the loss of lipase activity. Our study demonstrates the adiposome as a powerful, manipulatable model system that mimics the function of LD for binding and enzymatic activity studies of LD proteins in vitro.
Collapse
Affiliation(s)
- Xuejing Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zelun Zhi
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, Australia
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chang Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Adam Mechler
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, Australia
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
30
|
Monson EA, Trenerry AM, Laws JL, Mackenzie JM, Helbig KJ. Lipid droplets and lipid mediators in viral infection and immunity. FEMS Microbiol Rev 2021; 45:fuaa066. [PMID: 33512504 PMCID: PMC8371277 DOI: 10.1093/femsre/fuaa066] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Lipid droplets (LDs) contribute to key pathways important for the physiology and pathophysiology of cells. In a homeostatic view, LDs regulate the storage of neutral lipids, protein sequestration, removal of toxic lipids and cellular communication; however, recent advancements in the field show these organelles as essential for various cellular stress response mechanisms, including inflammation and immunity, with LDs acting as hubs that integrate metabolic and inflammatory processes. The accumulation of LDs has become a hallmark of infection, and is often thought to be virally driven; however, recent evidence is pointing to a role for the upregulation of LDs in the production of a successful immune response to viral infection. The fatty acids housed in LDs are also gaining interest due to the role that these lipid species play during viral infection, and their link to the synthesis of bioactive lipid mediators that have been found to have a very complex role in viral infection. This review explores the role of LDs and their subsequent lipid mediators during viral infections and poses a paradigm shift in thinking in the field, whereby LDs may play pivotal roles in protecting the host against viral infection.
Collapse
Affiliation(s)
- Ebony A Monson
- School of Life Sciences, La Trobe University, Melbourne, Australia, 3083
| | - Alice M Trenerry
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia, 3000
| | - Jay L Laws
- School of Life Sciences, La Trobe University, Melbourne, Australia, 3083
| | - Jason M Mackenzie
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia, 3000
| | - Karla J Helbig
- School of Life Sciences, La Trobe University, Melbourne, Australia, 3083
| |
Collapse
|
31
|
Abstract
Lipid droplets (LDs) are endoplasmic reticulum-derived organelles that consist of a core of neutral lipids encircled by a phospholipid monolayer decorated with proteins. As hubs of cellular lipid and energy metabolism, LDs are inherently involved in the etiology of prevalent metabolic diseases such as obesity and nonalcoholic fatty liver disease. The functions of LDs are regulated by a unique set of associated proteins, the LD proteome, which includes integral membrane and peripheral proteins. These proteins control key activities of LDs such as triacylglycerol synthesis and breakdown, nutrient sensing and signal integration, and interactions with other organelles. Here we review the mechanisms that regulate the composition of the LD proteome, such as pathways that mediate selective and bulk LD protein degradation and potential connections between LDs and cellular protein quality control.
Collapse
Affiliation(s)
- Melissa A Roberts
- Department of Molecular and Cell Biology and Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA;
| | - James A Olzmann
- Department of Molecular and Cell Biology and Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California 94720, USA; .,Chan Zuckerberg Biohub, San Francisco, California 94158, USA
| |
Collapse
|
32
|
Hussain SS, Tran TM, Ware TB, Luse MA, Prevost CT, Ferguson AN, Kashatus JA, Hsu KL, Kashatus DF. RalA and PLD1 promote lipid droplet growth in response to nutrient withdrawal. Cell Rep 2021; 36:109451. [PMID: 34320341 PMCID: PMC8344381 DOI: 10.1016/j.celrep.2021.109451] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 06/04/2021] [Accepted: 07/02/2021] [Indexed: 01/22/2023] Open
Abstract
Lipid droplets (LDs) are dynamic organelles that undergo dynamic changes in response to changing cellular conditions. During nutrient depletion, LD numbers increase to protect cells against toxic fatty acids generated through autophagy and provide fuel for beta-oxidation. However, the precise mechanisms through which these changes are regulated have remained unclear. Here, we show that the small GTPase RalA acts downstream of autophagy to directly facilitate LD growth during nutrient depletion. Mechanistically, RalA performs this function through phospholipase D1 (PLD1), an enzyme that converts phosphatidylcholine (PC) to phosphatidic acid (PA) and that is recruited to lysosomes during nutrient stress in a RalA-dependent fashion. RalA inhibition prevents recruitment of the LD-associated protein perilipin 3, which is required for LD growth. Our data support a model in which RalA recruits PLD1 to lysosomes during nutrient deprivation to promote the localized production of PA and the recruitment of perilipin 3 to expanding LDs.
Collapse
Affiliation(s)
- Syed S Hussain
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Tuyet-Minh Tran
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Timothy B Ware
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904, USA
| | - Melissa A Luse
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Christopher T Prevost
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Ashley N Ferguson
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Jennifer A Kashatus
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA
| | - Ku-Lung Hsu
- Department of Chemistry, University of Virginia, Charlottesville, VA 22904, USA; University of Virginia Cancer Center, University of Virginia Health System, Charlottesville, VA 22903, USA
| | - David F Kashatus
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA 22908, USA; University of Virginia Cancer Center, University of Virginia Health System, Charlottesville, VA 22903, USA.
| |
Collapse
|
33
|
Brink JTR, Fourie R, Sebolai O, Albertyn J, Pohl CH. The role of lipid droplets in microbial pathogenesis. J Med Microbiol 2021; 70. [PMID: 34184983 DOI: 10.1099/jmm.0.001383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The nonpolar lipids present in cells are mainly triacylglycerols and steryl esters. When cells are provided with an abundance of nutrients, these storage lipids accumulate. As large quantities of nonpolar lipids cannot be integrated into membranes, they are isolated from the cytosolic environment in lipid droplets. As specialized, inducible cytoplasmic organelles, lipid droplets have functions beyond the regulation of lipid metabolism, in cell signalling and activation, membrane trafficking and control of inflammatory mediator synthesis and secretion. Pathogens, including fungi, viruses, parasites, or intracellular bacteria can induce and may benefit from lipid droplets in infected cells. Here we review biogenesis of lipid droplets as well as the role of lipid droplets in the pathogenesis of selected viruses, bacteria, protists and yeasts.
Collapse
Affiliation(s)
- Jacobus T R Brink
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Ruan Fourie
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Olihile Sebolai
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Jacobus Albertyn
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Carolina H Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
34
|
Deng Y, Zhou C, Mirza AH, Bamigbade AT, Zhang S, Xu S, Liu P. Rab18 binds PLIN2 and ACSL3 to mediate lipid droplet dynamics. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158923. [PMID: 33713834 DOI: 10.1016/j.bbalip.2021.158923] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/26/2021] [Accepted: 03/05/2021] [Indexed: 01/16/2023]
Abstract
Lipid droplet (LD) is a vital organelle governing lipid homeostasis and Rab18 has been linked to lipid metabolism. However, the mechanisms of Rab18-mediated LD dynamics in myoblast cells remain elusive. Here, we report that Rab18 plays an important role in oleic acid (OA)-induced LD accumulation in mouse myoblast C2C12 cells. Rab18 was translocated from the endoplasmic reticulum (ER) to LDs during LD accumulation, which was regulated by perilipin 2 (PLIN2), a major LD protein. LD-associated Rab18 bound with the C terminus of PLIN2 and the LD localization of Rab18 was diminished when PLIN2 was depleted. Moreover, loss of function of Rab18 led to reduced triacylglycerol (TAG) level and fewer but larger LDs. In contrast, overexpression of Rab18 resulted in elevated TAG content and LD number. Furthermore, LD-associated Rab18 interacted with acyl-CoA synthetase long-chain family member 3 (ACSL3), which in turn promoted the LD localization of this protein. These data show that Rab18 interacts with PLIN2 and forms a complex with PLIN2 and ACSL3, which plays a critical role in LD accumulation and dynamics of myoblast cells.
Collapse
Affiliation(s)
- Yaqin Deng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chang Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ahmed Hammad Mirza
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Adekunle T Bamigbade
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
35
|
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: 6] [Impact Index Per Article: 2.0] [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
|
36
|
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
|
37
|
Mirza AH, Cui L, Zhang S, Liu P. Comparative proteomics reveals that lipid droplet-anchored mitochondria are more sensitive to cold in brown adipocytes. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158992. [PMID: 34147658 DOI: 10.1016/j.bbalip.2021.158992] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 10/21/2022]
Abstract
Brown adipose tissue (BAT) is specialized for uncoupled heat production through mitochondrion fueled majorly from fatty acids (FAs) of lipid droplets (LDs). How the interaction between the two organelles contributes the generation of heat remains elusive. Here, we report that LD-anchored mitochondria (LDAM) were observed in the BAT of mice raised at three different temperatures, 30 °C, 23 °C, and 6 °C. The biochemical analyses including Western blotting of electron transport chain subunits showed that LDAM were functional. Comparative proteomics analysis was conducted, which revealed differential expressions of proteins between LDAM and cytoplasmic mitochondria (CM) at different temperatures. Higher expressions of proteins at low temperature were observed for i) FA β-oxidation in LDAM including FA synthesis and uncoupling, ii) pseudo-futile cycle in CM, and iii) two shuttle systems: glycerol 3-phosphate in both CM and LDAM and citrate malate in CM. Together, these results suggest that LDs and LDAM form a preorganized and functional organelle complex that permits the rapid response to cold.
Collapse
Affiliation(s)
- Ahmed Hammad Mirza
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liujuan Cui
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, China.
| |
Collapse
|
38
|
Formulative Study and Intracellular Fate Evaluation of Ethosomes and Transethosomes for Vitamin D3 Delivery. Int J Mol Sci 2021; 22:ijms22105341. [PMID: 34069489 PMCID: PMC8161393 DOI: 10.3390/ijms22105341] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
In this pilot study, ethosomes and transethosomes were investigated as potential delivery systems for cholecalciferol (vitamin D3), whose deficiency has been correlated to many disorders such as dermatological diseases, systemic infections, cancer and sarcopenia. A formulative study on the influence of pharmaceutically acceptable ionic and non-ionic surfactants allowed the preparation of different transethosomes. In vitro cytotoxicity was evaluated in different cell types representative of epithelial, connective and muscle tissue. Then, the selected nanocarriers were further investigated at light and transmission electron microscopy to evaluate their uptake and intracellular fate. Both ethosomes and transethosomes proven to have physicochemical properties optimal for transdermal penetration and efficient vitamin D3 loading; moreover, nanocarriers were easily internalized by all cell types, although they followed distinct intracellular fates: ethosomes persisted for long times inside the cytoplasm, without inducing subcellular alteration, while transethosomes underwent rapid degradation giving rise to an intracellular accumulation of lipids. These basic results provide a solid scientific background to in vivo investigations aimed at exploring the efficacy of vitamin D3 transdermal administration in different experimental and pathological conditions.
Collapse
|
39
|
Veerabagu M, Rinne PLH, Skaugen M, Paul LK, van der Schoot C. Lipid Body Dynamics in Shoot Meristems: Production, Enlargement, and Putative Organellar Interactions and Plasmodesmal Targeting. FRONTIERS IN PLANT SCIENCE 2021; 12:674031. [PMID: 34367200 PMCID: PMC8335594 DOI: 10.3389/fpls.2021.674031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/14/2021] [Indexed: 05/20/2023]
Abstract
Post-embryonic cells contain minute lipid bodies (LBs) that are transient, mobile, engage in organellar interactions, and target plasmodesmata (PD). While LBs can deliver γ-clade 1,3-β-glucanases to PD, the nature of other cargo is elusive. To gain insight into the poorly understood role of LBs in meristems, we investigated their dynamics by microscopy, gene expression analyzes, and proteomics. In developing buds, meristems accumulated LBs, upregulated several LB-specific OLEOSIN genes and produced OLEOSINs. During bud maturation, the major gene OLE6 was strongly downregulated, OLEOSINs disappeared from bud extracts, whereas lipid biosynthesis genes were upregulated, and LBs were enlarged. Proteomic analyses of the LB fraction of dormant buds confirmed that OLEOSINs were no longer present. Instead, we identified the LB-associated proteins CALEOSIN (CLO1), Oil Body Lipase 1 (OBL1), Lipid Droplet Interacting Protein (LDIP), Lipid Droplet Associated Protein1a/b (LDAP1a/b) and LDAP3a/b, and crucial components of the OLEOSIN-deubiquitinating and degradation machinery, such as PUX10 and CDC48A. All mRFP-tagged LDAPs localized to LBs when transiently expressed in Nicotiana benthamiana. Together with gene expression analyzes, this suggests that during bud maturation, OLEOSINs were replaced by LDIP/LDAPs at enlarging LBs. The LB fraction contained the meristem-related actin7 (ACT7), "myosin XI tail-binding" RAB GTPase C2A, an LB/PD-associated γ-clade 1,3-β-glucanase, and various organelle- and/or PD-localized proteins. The results are congruent with a model in which LBs, motorized by myosin XI-k/1/2, traffic on F-actin, transiently interact with other organelles, and deliver a diverse cargo to PD.
Collapse
Affiliation(s)
- Manikandan Veerabagu
- Faculty of Biosciences, Department of Plant Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Päivi L. H. Rinne
- Faculty of Biosciences, Department of Plant Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Morten Skaugen
- Faculty of Chemistry, Biotechnology, and Food Science, Norwegian University of Life Sciences, Ås, Norway
| | - Laju K. Paul
- Faculty of Biosciences, Department of Plant Sciences, Norwegian University of Life Sciences, Ås, Norway
| | - Christiaan van der Schoot
- Faculty of Biosciences, Department of Plant Sciences, Norwegian University of Life Sciences, Ås, Norway
- *Correspondence: Christiaan van der Schoot
| |
Collapse
|
40
|
Cui L, Liu P. Two Types of Contact Between Lipid Droplets and Mitochondria. Front Cell Dev Biol 2020; 8:618322. [PMID: 33385001 PMCID: PMC7769837 DOI: 10.3389/fcell.2020.618322] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022] Open
Abstract
Lipid droplets (LDs) and mitochondria are essential organelles involved in cellular lipid metabolism and energy homeostasis. Accumulated studies have revealed that the physical contact between these two organelles is important for their functions. Current understanding of the contact between cellular organelles is highly dynamic, fitting a "kiss-and-run" model. The same pattern of contact between LDs and mitochondria has been reported and several proteins are found to mediate this contact, such as perilipin1 (PLIN1) and PLIN5. Another format of the contact has also been found and termed anchoring. LD-anchored mitochondria (LDAM) are identified in oxidative tissues including brown adipose tissue (BAT), skeletal muscle, and heart muscle, and this anchoring between these two organelles is conserved from mouse to monkey. Moreover, this anchoring is generated during the brown/beige adipocyte differentiation. In this review, we will summarize previous studies on the interaction between LDs and mitochondria, categorize the types of the contacts into dynamic and stable/anchored, present their similarities and differences, discuss their potential distinct molecular mechanism, and finally propose a working hypothesis that may explain why and how cells use two patterns of contact between LDs and mitochondria.
Collapse
Affiliation(s)
- Liujuan Cui
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
41
|
Leyland B, Zarka A, Didi-Cohen S, Boussiba S, Khozin-Goldberg I. High Resolution Proteome of Lipid Droplets Isolated from the Pennate Diatom Phaeodactylum tricornutum (Bacillariophyceae) Strain pt4 provides mechanistic insights into complex intracellular coordination during nitrogen deprivation. JOURNAL OF PHYCOLOGY 2020; 56:1642-1663. [PMID: 32779202 DOI: 10.1111/jpy.13063] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/14/2020] [Accepted: 07/12/2020] [Indexed: 05/08/2023]
Abstract
Lipid droplets (LDs) are an organelle conserved amongst all eukaryotes, consisting of a neutral lipid core surrounded by a polar lipid monolayer. Many species of microalgae accumulate LDs in response to stress conditions, such as nitrogen starvation. Here, we report the isolation and proteomic profiling of LD proteins from the model oleaginous pennate diatom Phaeodactylum tricornutum, strain Pt4 (UTEX 646). We also provide a quantitative description of LD morphological ontogeny, and fatty acid content. Novel cell disruption and LD isolation methods, combined with suspension-trapping and nanoflow liquid chromatography coupled to high resolution mass spectrometry, yielded an unprecedented number of LD proteins. Predictive annotation of the LD proteome suggests a broad assemblage of proteins with diverse functions, including lipid metabolism and vesicle trafficking, as well as ribosomal and proteasomal machinery. These proteins provide mechanistic insights into LD processes, and evidence for interactions between LDs and other organelles. We identify for the first time several key steps in diatom LD-associated triacylglycerol biosynthesis. Bioinformatic analyses of the LD proteome suggests multiple protein targeting mechanisms, including amphipathic helices, post-translational modifications, and translocation machinery. This work corroborates recent findings from other strains of P. tricornutum, other diatoms, and other eukaryotic organisms, suggesting that the fundamental proteins orchestrating LDs are conserved, and represent an ancient component of the eukaryotic endomembrane system. We postulate a comprehensive model of nitrogen starvation-induced diatom LDs on a molecular scale, and provide a wealth of candidates for metabolic engineering, with the potential to eventually customize LD contents.
Collapse
Affiliation(s)
- Ben Leyland
- The Microalgal Biotechnology Laboratory, The French Associates Institute for Agriculture and Biotechnology, Jacob Blaustein Institute for Desert Research, Ben-Gurion University of the Negev, Sede Boker Campus, Be'er Sheva, 84990, Israel
| | - Aliza Zarka
- The Microalgal Biotechnology Laboratory, The French Associates Institute for Agriculture and Biotechnology, Jacob Blaustein Institute for Desert Research, Ben-Gurion University of the Negev, Sede Boker Campus, Be'er Sheva, 84990, Israel
| | - Shoshana Didi-Cohen
- The Microalgal Biotechnology Laboratory, The French Associates Institute for Agriculture and Biotechnology, Jacob Blaustein Institute for Desert Research, Ben-Gurion University of the Negev, Sede Boker Campus, Be'er Sheva, 84990, Israel
| | - Sammy Boussiba
- The Microalgal Biotechnology Laboratory, The French Associates Institute for Agriculture and Biotechnology, Jacob Blaustein Institute for Desert Research, Ben-Gurion University of the Negev, Sede Boker Campus, Be'er Sheva, 84990, Israel
| | - Inna Khozin-Goldberg
- The Microalgal Biotechnology Laboratory, The French Associates Institute for Agriculture and Biotechnology, Jacob Blaustein Institute for Desert Research, Ben-Gurion University of the Negev, Sede Boker Campus, Be'er Sheva, 84990, Israel
| |
Collapse
|
42
|
Sun J, Xu X, Huang X, Ji S, Bian C, Ji H. Nuclear factor-κB subunit p65 is involved in lipopolysaccharide-induced lipid accumulation via regulating DGAT1b in Ctenopharyngodon idellus kidney cells. FISH & SHELLFISH IMMUNOLOGY 2020; 105:71-77. [PMID: 32585360 DOI: 10.1016/j.fsi.2020.05.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 06/11/2023]
Abstract
Lipopolysaccharide (LPS) can promote the accumulation of triglycerides (TGs) in CIK (Ctenopharyngodon idellus kidney) cells, but the underlying mechanism is unclear. In this study, two genes involved TG synthesis, DGAT1a and DGAT1b, were isolated and characterized from grass carp Ctenopharyngodon idella, which encode peptides of 498 and 501 amino acids, respectively. Phylogenetic and synteny analyses indicated that DGAT1a and DGAT1b could have originated from the teleost-specific genome duplication event. Analysis of the exon-intron structures clarified that genomic structures of all DGAT1 proteins are conserved in vertebrates. DGAT1a mRNA was highly expressed in gut, adipose tissue and heart, while DGAT1b mRNA was highly expressed in liver and kidney. After LPS treatment, only expression of DGAT1b was up-regulated and knockdown of DGAT1b reduced the content of TG, suggesting that DGAT1b is involved in LPS-induced lipid accumulation. To explore the mechanism underlying the transcriptional regulation of DGAT1b in response to LPS, we cloned DGAT1b promoter sequence. Its promoter sequence consists of IRF7, RelA (p65) and RelB binding elements. Dual luciferase assay and q-PCR suggested that the promoter of DGAT1b can be activated by the overexpression of p65, but cannot be triggered by IRF7 and RelB. Mutational analysis shows that the potential p65 binding sites may locate in the region -111/-100 bp of the DGAT1b promoter. These results indicated that DGAT1b is the target gene of NF-κB p65. Finally, inhibiting p65 effectively decreased LPS-induced lipid accumulation. Taken together, we demonstrate that NF-κB p65 takes part in the lipid accumulation by regulating DGAT1b-induced TG synthesis in LPS signalling in CIK cells. The finding that NF-κB p65 links LPS signalling and TG synthesis adds to our growing appreciation of the interplay between immunity and lipid metabolism in fish.
Collapse
Affiliation(s)
- Jian Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xinxin Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaocheng Huang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Shanghong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chenchen Bian
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
43
|
Ali AS, Chen R, Raju R, Kshirsagar R, Gilbert A, Zang L, Karger BL, Ivanov AR. Multi-Omics Reveals Impact of Cysteine Feed Concentration and Resulting Redox Imbalance on Cellular Energy Metabolism and Specific Productivity in CHO Cell Bioprocessing. Biotechnol J 2020; 15:e1900565. [PMID: 32170810 PMCID: PMC7880547 DOI: 10.1002/biot.201900565] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/18/2020] [Indexed: 12/16/2022]
Abstract
Chinese hamster ovary (CHO) cells are currently the primary host cell lines used in biotherapeutic manufacturing of monoclonal antibodies (mAbs) and other biopharmaceuticals. Cellular energy metabolism and endoplasmic reticulum (ER) stress are known to greatly impact cell growth, viability, and specific productivity of a biotherapeutic; but the molecular mechanisms are not fully understood. The authors previously employed multi-omics profiling to investigate the impact of a reduction in cysteine (Cys) feed concentration in a fed-batch process and found that disruption of the redox balance led to a substantial decline in cell viability and titer. Here, the multi-omics findings are expanded, and the impact redox imbalance has on ER stress, mitochondrial homeostasis, and lipid metabolism is explored. The reduced Cys feed activates the amino acid response (AAR), increases mitochondrial stress, and initiates gluconeogenesis. Multi-omics analysis reveals that together, ER stress and AAR signaling shift the cellular energy metabolism to rely primarily on anaplerotic reactions, consuming amino acids and producing lactate, to maintain energy generation. Furthermore, the pathways are demonstrated in which this shift in metabolism leads to a substantial decline in specific productivity and altered mAb glycosylation. Through this work, meaningful bioprocess markers and targets for genetic engineering are identified.
Collapse
Affiliation(s)
- Amr S Ali
- Cell Culture Development, Biogen Inc., Cambridge, MA, 02142, USA
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, USA
- Analytical Development, Biogen Inc., Cambridge, MA, 02142, USA
| | - Rachel Chen
- Analytical Development, Biogen Inc., Cambridge, MA, 02142, USA
| | - Ravali Raju
- Cell Culture Development, Biogen Inc., Cambridge, MA, 02142, USA
| | | | - Alan Gilbert
- Cell Culture Development, Biogen Inc., Cambridge, MA, 02142, USA
| | - Li Zang
- Analytical Development, Biogen Inc., Cambridge, MA, 02142, USA
| | - Barry L Karger
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, USA
| | - Alexander R Ivanov
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
44
|
Li Z, Weller SG, Drizyte-Miller K, Chen J, Krueger EW, Mehall B, Stöckli J, Casey CA, Cao H, McNiven MA. Maturation of Lipophagic Organelles in Hepatocytes Is Dependent Upon a Rab10/Dynamin-2 Complex. Hepatology 2020; 72:486-502. [PMID: 31808574 PMCID: PMC8919976 DOI: 10.1002/hep.31059] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/14/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS Hepatocytes play a central role in storage and utilization of fat by the liver. Selective breakdown of lipid droplets (LDs) by autophagy (also called lipophagy) is a key process utilized to catabolize these lipids as an energy source. How the autophagic machinery is selectively targeted to LDs, where it mediates membrane engulfment and subsequent degradation, is unclear. Recently, we have reported that two distinct GTPases, the mechanoenzyme, dynamin2 (Dyn2), and the small regulatory Rab GTPase, Rab10, work independently at distinct steps of lipophagy in hepatocytes. APPROACH AND RESULTS In an attempt to understand how these proteins are regulated and recruited to autophagic organelles, we performed a nonbiased biochemical screen for Dyn2-binding partners and found that Dyn2 actually binds Rab10 directly through a defined effector domain of Rab10 and the middle domain of Dyn2. These two GTPases can be observed to interact transiently on membrane tubules in hepatoma cells and along LD-centric autophagic membranes. Most important, we found that a targeted disruption of this interaction leads to an inability of cells to trim tubulated cytoplasmic membranes, some of which extend from lipophagic organelles, resulting in LD accumulation. CONCLUSIONS This study identifies a functional, and direct, interaction between Dyn2 and a regulatory Rab GTPase that may play an important role in hepatocellular metabolism.
Collapse
Affiliation(s)
- Zhipeng Li
- Biochemistry and Molecular Biology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN
| | - Shaun G. Weller
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Kristina Drizyte-Miller
- Biochemistry and Molecular Biology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN
| | - Jing Chen
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Eugene W. Krueger
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Bridget Mehall
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Jacqueline Stöckli
- Charles Perkins Centre, School of Life and Environmental Sciences, University of Sydney, Sydney, Australia
| | - Carol A. Casey
- Department of Internal Medicine, University of Nebraska Medical Center, 988090 Nebraska Medical Center, Omaha, NE
| | - Hong Cao
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN
| | - Mark A. McNiven
- Biochemistry and Molecular Biology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN,Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| |
Collapse
|
45
|
Chua NK, Coates HW, Brown AJ. Squalene monooxygenase: a journey to the heart of cholesterol synthesis. Prog Lipid Res 2020; 79:101033. [DOI: 10.1016/j.plipres.2020.101033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/21/2020] [Accepted: 04/24/2020] [Indexed: 02/07/2023]
|
46
|
Coleman RA. The "discovery" of lipid droplets: A brief history of organelles hidden in plain sight. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158762. [PMID: 32622088 DOI: 10.1016/j.bbalip.2020.158762] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022]
Abstract
Mammalian lipid droplets (LDs), first described as early as the 1880s, were virtually ignored for more than 100 years. Between 1991 and the early 2000s, however, a series of discoveries and conceptual breakthroughs led to a resurgent interest in obesity as a disease, in the metabolism of intracellular triacylglycerol (TAG), and in the physical locations of LDs as cellular structures with their associated proteins. Insights included the recognition that obesity underlies major chronic diseases, that appetite is hormonally controlled, that hepatic steatosis is not a benign finding, and that diabetes might fundamentally be a disorder of lipid metabolism. In this brief review, I describe the metamorphosis of LDs from overlooked globs of stored fat to dynamic organelles that control insulin resistance, mitochondrial oxidation, and viral replication.
Collapse
Affiliation(s)
- Rosalind A Coleman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, United States of America.
| |
Collapse
|
47
|
|
48
|
Lundquist PK, Shivaiah KK, Espinoza-Corral R. Lipid droplets throughout the evolutionary tree. Prog Lipid Res 2020; 78:101029. [PMID: 32348789 DOI: 10.1016/j.plipres.2020.101029] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 04/11/2020] [Accepted: 04/18/2020] [Indexed: 12/11/2022]
Abstract
Intracellular lipid droplets are utilized for lipid storage and metabolism in organisms as evolutionarily diverse as animals, fungi, plants, bacteria, and archaea. These lipid droplets demonstrate great diversity in biological functions and protein and lipid compositions, yet fundamentally share common molecular and ultrastructural characteristics. Lipid droplet research has been largely fragmented across the diversity of lipid droplet classes and sub-classes. However, we suggest that there is great potential benefit to the lipid community in better integrating the lipid droplet research fields. To facilitate such integration, we survey the protein and lipid compositions, functional roles, and mechanisms of biogenesis across the breadth of lipid droplets studied throughout the natural world. We depict the big picture of lipid droplet biology, emphasizing shared characteristics and unique differences seen between different classes. In presenting the known diversity of lipid droplets side-by-side it becomes necessary to offer for the first time a consistent system of categorization and nomenclature. We propose a division into three primary classes that reflect their sub-cellular location: i) cytoplasmic lipid droplets (CYTO-LDs), that are present in the eukaryotic cytoplasm, ii) prokaryotic lipid droplets (PRO-LDs), that exist in the prokaryotic cytoplasm, and iii) plastid lipid droplets (PL-LDs), that are found in plant plastids, organelles of photosynthetic eukaryotes. Within each class there is a remarkable array of sub-classes displaying various sizes, shapes and compositions. A more integrated lipid droplet research field will provide opportunities to better build on discoveries and accelerate the pace of research in ways that have not been possible.
Collapse
Affiliation(s)
- Peter K Lundquist
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Plant Resilience Institute, Michigan State University, East Lansing, MI, 48824, USA.
| | - Kiran-Kumar Shivaiah
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Plant Resilience Institute, Michigan State University, East Lansing, MI, 48824, USA
| | - Roberto Espinoza-Corral
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI 48824, USA; Plant Resilience Institute, Michigan State University, East Lansing, MI, 48824, USA
| |
Collapse
|
49
|
Mejhert N, Kuruvilla L, Gabriel KR, Elliott SD, Guie MA, Wang H, Lai ZW, Lane EA, Christiano R, Danial NN, Farese RV, Walther TC. Partitioning of MLX-Family Transcription Factors to Lipid Droplets Regulates Metabolic Gene Expression. Mol Cell 2020; 77:1251-1264.e9. [PMID: 32023484 PMCID: PMC7397554 DOI: 10.1016/j.molcel.2020.01.014] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/05/2019] [Accepted: 01/07/2020] [Indexed: 12/22/2022]
Abstract
Lipid droplets (LDs) store lipids for energy and are central to cellular lipid homeostasis. The mechanisms coordinating lipid storage in LDs with cellular metabolism are unclear but relevant to obesity-related diseases. Here we utilized genome-wide screening to identify genes that modulate lipid storage in macrophages, a cell type involved in metabolic diseases. Among ∼550 identified screen hits is MLX, a basic helix-loop-helix leucine-zipper transcription factor that regulates metabolic processes. We show that MLX and glucose-sensing family members MLXIP/MondoA and MLXIPL/ChREBP bind LDs via C-terminal amphipathic helices. When LDs accumulate in cells, these transcription factors bind to LDs, reducing their availability for transcriptional activity and attenuating the response to glucose. Conversely, the absence of LDs results in hyperactivation of MLX target genes. Our findings uncover a paradigm for a lipid storage response in which binding of MLX transcription factors to LD surfaces adjusts the expression of metabolic genes to lipid storage levels.
Collapse
Affiliation(s)
- Niklas Mejhert
- 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
| | - Leena Kuruvilla
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Katlyn R Gabriel
- 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
| | - Shane D Elliott
- 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
| | - Marie-Aude Guie
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Huajin Wang
- Department of Cell Biology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Zon Weng Lai
- 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
| | - Elizabeth A Lane
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Romain Christiano
- 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
| | - Nika N Danial
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, 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 MIT and Harvard, Cambridge, MA 02142, 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 MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
50
|
Leyland B, Boussiba S, Khozin-Goldberg I. A Review of Diatom Lipid Droplets. BIOLOGY 2020; 9:biology9020038. [PMID: 32098118 PMCID: PMC7168155 DOI: 10.3390/biology9020038] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/12/2020] [Accepted: 02/14/2020] [Indexed: 12/20/2022]
Abstract
The dynamic nutrient availability and photon flux density of diatom habitats necessitate buffering capabilities in order to maintain metabolic homeostasis. This is accomplished by the biosynthesis and turnover of storage lipids, which are sequestered in lipid droplets (LDs). LDs are an organelle conserved among eukaryotes, composed of a neutral lipid core surrounded by a polar lipid monolayer. LDs shield the intracellular environment from the accumulation of hydrophobic compounds and function as a carbon and electron sink. These functions are implemented by interconnections with other intracellular systems, including photosynthesis and autophagy. Since diatom lipid production may be a promising objective for biotechnological exploitation, a deeper understanding of LDs may offer targets for metabolic engineering. In this review, we provide an overview of diatom LD biology and biotechnological potential.
Collapse
|