1
|
Chen PHB, Li XL, Baskin JM. Synthetic Lipid Biology. Chem Rev 2025; 125:2502-2560. [PMID: 39805091 PMCID: PMC11969270 DOI: 10.1021/acs.chemrev.4c00761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Cells contain thousands of different lipids. Their rapid and redundant metabolism, dynamic movement, and many interactions with other biomolecules have justly earned lipids a reputation as a vexing class of molecules to understand. Further, as the cell's hydrophobic metabolites, lipids assemble into supramolecular structures─most commonly bilayers, or membranes─from which they carry out myriad biological functions. Motivated by this daunting complexity, researchers across disciplines are bringing order to the seeming chaos of biological lipids and membranes. Here, we formalize these efforts as "synthetic lipid biology". Inspired by the idea, central to synthetic biology, that our abilities to understand and build biological systems are intimately connected, we organize studies and approaches across numerous fields to create, manipulate, and analyze lipids and biomembranes. These include construction of lipids and membranes from scratch using chemical and chemoenzymatic synthesis, editing of pre-existing membranes using optogenetics and protein engineering, detection of lipid metabolism and transport using bioorthogonal chemistry, and probing of lipid-protein interactions and membrane biophysical properties. What emerges is a portrait of an incipient field where chemists, biologists, physicists, and engineers work together in proximity─like lipids themselves─to build a clearer description of the properties, behaviors, and functions of lipids and membranes.
Collapse
Affiliation(s)
- Po-Hsun Brian Chen
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Xiang-Ling Li
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| | - Jeremy M Baskin
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, New York 14853, United States
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, United States
| |
Collapse
|
2
|
Holzhütter H. Dynamical modelling of lipid droplet formation suggests a key function of membrane phospholipids. FEBS J 2025; 292:206-225. [PMID: 39132700 PMCID: PMC11705222 DOI: 10.1111/febs.17238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/10/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024]
Abstract
Cells store triacylglycerol (TAG) within lipid droplets (LDs). A dynamic model describing complete LD formation at the endoplasmic reticulum (ER) membrane does not yet exist. A biochemical-biophysical model of LD synthesis is proposed. It describes the time-dependent accumulation of TAG in the ER membrane as the formation of a potential LD (pLD) bounded by spherical caps of the inner and outer monolayers of the membrane. The expansion rate of the pLD depends on the TAG supply, the elastic properties of the ER membrane, and the recruitment of phospholipids (PLs) to the cap-covering monolayers. Model simulations provided the following insights: (a) Marginal differences in the surface tension of the cap monolayers are sufficient to fully drive the expansion of the pLD towards the cytosol or lumen. (b) Selective reduction of PL supply to the luminal monolayer ensures stable formation of cytosolic LDs, irrespective of variations in the elasto-mechanical properties of the ER membrane. (c) The rate of TAG supply to the cytosolic monolayer has a major effect on the size and maturation time of LDs but has no significant effect on the TAG export per individual LD. The recruitment of additional PLs to the cap monolayers of pLDs critically controls the budding direction, size, and maturation time of LDs. The ability of cells to acquire additional LD initiation sites appears to be key to coping with acutely high levels of potentially toxic free fatty acids.
Collapse
Affiliation(s)
- Hermann‐Georg Holzhütter
- Institute of BiochemistryCharité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinGermany
| |
Collapse
|
3
|
Prakash P, Manchanda P, Paouri E, Bisht K, Sharma K, Rajpoot J, Wendt V, Hossain A, Wijewardhane PR, Randolph CE, Chen Y, Stanko S, Gasmi N, Gjojdeshi A, Card S, Fine J, Jethava KP, Clark MG, Dong B, Ma S, Crockett A, Thayer EA, Nicolas M, Davis R, Hardikar D, Allende D, Prayson RA, Zhang C, Davalos D, Chopra G. Amyloid β Induces Lipid Droplet-Mediated Microglial Dysfunction in Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.04.543525. [PMID: 37333071 PMCID: PMC10274698 DOI: 10.1101/2023.06.04.543525] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Several microglia-expressed genes have emerged as top risk variants for Alzheimer's disease (AD). Impaired microglial phagocytosis is one of the main proposed outcomes by which these AD-risk genes may contribute to neurodegeneration, but the mechanisms translating genetic association to cellular dysfunction remain unknown. Here we show that microglia form lipid droplets (LDs) upon exposure to amyloid-beta (Aβ), and that their LD load increases with proximity to amyloid plaques in brains from human patients and the AD mouse model 5xFAD. LD formation is dependent on age and disease progression and is prominent in the hippocampus in mice and humans. Despite differences in microglial LD load between brain regions and sexes in mice, LD-laden microglia exhibited a deficit in Aβ phagocytosis. Unbiased lipidomic analysis identified a decrease in free fatty acids (FFAs) and a parallel increase in triacylglycerols (TGs) as the key metabolic transition underlying LD formation. DGAT2, a key enzyme for converting FFAs to TGs, promotes microglial LD formation and is increased in 5xFAD and human AD brains. Inhibition or degradation of DGAT2 improved microglial uptake of Aβ and drastically reduced plaque load in 5xFAD mice, respectively. These findings identify a new lipid-mediated mechanism underlying microglial dysfunction that could become a novel therapeutic target for AD.
Collapse
Affiliation(s)
- Priya Prakash
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Palak Manchanda
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Evi Paouri
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Kanchan Bisht
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Kaushik Sharma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Jitika Rajpoot
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Victoria Wendt
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Ahad Hossain
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | | | | | - Yihao Chen
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Sarah Stanko
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Nadia Gasmi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Anxhela Gjojdeshi
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Sophie Card
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Jonathan Fine
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Krupal P. Jethava
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Matthew G. Clark
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Bin Dong
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Seohee Ma
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Alexis Crockett
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | | | - Marlo Nicolas
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Ryann Davis
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dhruv Hardikar
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Daniela Allende
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Richard A. Prayson
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Chi Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Dimitrios Davalos
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case, Western Reserve University, Cleveland, OH 44106, USA
| | - Gaurav Chopra
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute for Drug Discovery, Purdue University, West Lafayette, IN 47907, USA
- Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
- Purdue Institute of Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
4
|
Zhang H, Fang J, Chu K, Smith ZJ. Real-Time Analysis of Lipid Droplet Morpho-Chemical Dynamics in Living Human Hepatocytes via Phase-Guided Raman Sampling. Anal Chem 2024. [PMID: 39566093 DOI: 10.1021/acs.analchem.4c03633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Lipid droplets (LDs) are highly dynamic organelles, undertaking many important functions such as maintaining lipid metabolism and cellular homeostasis. Traditional methods to analyze LD dynamics focus on morphological changes, while chemical dynamics cannot be easily probed with traditional analytical chemistry techniques. To overcome this challenge, we show here how our phase-guided Raman sampling method, where high-resolution phase microscopy images direct a Raman sampling beam, can perform label-free, multimodal characterization of LD dynamics in living cells at both the single-cell and single-LD levels with submicron accuracy and high temporal resolution. We demonstrate the study of the morphological-compositional dynamics of human hepatocellular carcinoma cells (PLC cells) under different environmental conditions and with and without fatty acid supplementation, providing insight into LD heterogeneity and heterogeneity of response. Finally, we introduce a measurement method for the dynamics of cell-average LD composition, which can quickly and accurately characterize the lipid dynamics at the single-cell level with <30 s temporal resolution. The results here show the promise of the phase-guided Raman sampling method for dynamic morpho-chemical profiling of organelle populations.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jingde Fang
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Kaiqin Chu
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou, Jiangsu 215123, China
| | - Zachary J Smith
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
5
|
Khan N, Mohd Salmi T, Karamalakis AP, Ramdas Nair A, Sadler KC, Cox AG. Optimized methods to image hepatic lipid droplets in zebrafish larvae. Dis Model Mech 2024; 17:dmm050786. [PMID: 39373223 PMCID: PMC11625896 DOI: 10.1242/dmm.050786] [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: 03/14/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024] Open
Abstract
The optical transparency of zebrafish larvae enables visualization of subcellular structures in intact organs, and these vertebrates are widely used to study lipid biology and liver disease. Lipid droplet (LD) presence is a prevalent feature of healthy cells, but, under conditions such as nutrient excess, toxicant exposure or metabolic imbalance, LD accumulation in hepatocytes can be a harbinger of more severe forms of liver disease. We undertook a comprehensive analysis of approaches useful to investigate LD distribution and dynamics in physiological and pathological conditions in the liver of zebrafish larvae. This comparative analysis of the lipid dyes Oil Red O, Nile Red, LipidTox and LipidSpot, as well as transgenic LD reporters that rely on EGFP fusions of the LD-decorating protein perilipin 2 (PLIN2), demonstrates the strengths and limitations of each approach. These protocols are amenable to detection methods ranging from low-resolution stereomicroscopy to confocal imaging, which enables measurements of hepatic LD size, number and dynamics at cellular resolution in live and fixed animals. This resource will benefit investigators studying LD biology in zebrafish disease models.
Collapse
Affiliation(s)
- Nouf Khan
- Program in Biology, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Talhah Mohd Salmi
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Anthony P. Karamalakis
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Anjana Ramdas Nair
- Program in Biology, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Kirsten C. Sadler
- Program in Biology, New York University Abu Dhabi, PO Box 129188, Abu Dhabi, United Arab Emirates
| | - Andrew G. Cox
- Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC 3000, Australia
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
6
|
Wang F, Huynh PM, An YA. Mitochondrial Function and Dysfunction in White Adipocytes and Therapeutic Implications. Compr Physiol 2024; 14:5581-5640. [PMID: 39382163 DOI: 10.1002/cphy.c230009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
For a long time, white adipocytes were thought to function as lipid storages due to the sizeable unilocular lipid droplet that occupies most of their space. However, recent discoveries have highlighted the critical role of white adipocytes in maintaining energy homeostasis and contributing to obesity and related metabolic diseases. These physiological and pathological functions depend heavily on the mitochondria that reside in white adipocytes. This article aims to provide an up-to-date overview of the recent research on the function and dysfunction of white adipocyte mitochondria. After briefly summarizing the fundamental aspects of mitochondrial biology, the article describes the protective role of functional mitochondria in white adipocyte and white adipose tissue health and various roles of dysfunctional mitochondria in unhealthy white adipocytes and obesity. Finally, the article emphasizes the importance of enhancing mitochondrial quantity and quality as a therapeutic avenue to correct mitochondrial dysfunction, promote white adipocyte browning, and ultimately improve obesity and its associated metabolic diseases. © 2024 American Physiological Society. Compr Physiol 14:5581-5640, 2024.
Collapse
Affiliation(s)
- Fenfen Wang
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Phu M Huynh
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| | - Yu A An
- Department of Anesthesiology, Critical Care, and Pain Medicine, Center for Perioperative Medicine, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Center for Metabolic and Degenerative Diseases, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
- Department of Biochemistry and Molecular Biology, McGovern Medical School, UT Health Science Center at Houston, Houston, Texas, USA
| |
Collapse
|
7
|
Farías MA, Diethelm-Varela B, Kalergis AM, González PA. Interplay between lipid metabolism, lipid droplets and RNA virus replication. Crit Rev Microbiol 2024; 50:515-539. [PMID: 37348003 DOI: 10.1080/1040841x.2023.2224424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 09/20/2022] [Accepted: 01/29/2023] [Indexed: 06/24/2023]
Abstract
Lipids play essential roles in the cell as components of cellular membranes, signaling molecules, and energy storage sources. Lipid droplets are cellular organelles composed of neutral lipids, such as triglycerides and cholesterol esters, and are also considered as cellular energy reserves, yet new functions have been recently associated with these structures, such as regulators of oxidative stress and cellular lipotoxicity, as well as modulators of pathogen infection through immune regulation. Lipid metabolism and lipid droplets participate in the infection process of many RNA viruses and control their replication and assembly, among others. Here, we review and discuss the contribution of lipid metabolism and lipid droplets over the replication cycle of RNA viruses, altogether pointing out potentially new pharmacological antiviral targets associated with lipid metabolism.
Collapse
Affiliation(s)
- Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Benjamín Diethelm-Varela
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
8
|
Gandhi SA, Parveen S, Alduhailan M, Tripathi R, Junedi N, Saqallah M, Sanders MA, Hoffmann PM, Truex K, Granneman JG, Kelly CV. Methods for making and observing model lipid droplets. CELL REPORTS METHODS 2024; 4:100774. [PMID: 38749444 PMCID: PMC11133809 DOI: 10.1016/j.crmeth.2024.100774] [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: 10/06/2023] [Revised: 01/30/2024] [Accepted: 04/19/2024] [Indexed: 05/23/2024]
Abstract
We present methods for making and testing the membrane biophysics of model lipid droplets (LDs). Methods are described for imaging LDs ranging in size from 0.1 to 40 μm in diameter with high-resolution microscopy and spectroscopy. With known LD compositions, membrane binding, sorting, diffusion, and tension were measured via fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), fluorescence lifetime imaging microscopy (FLIM), atomic force microscopy (AFM), and imaging flow cytometry. Additionally, a custom, small-volume pendant droplet tensiometer is described and used to measure the association of phospholipids to the LD surface. These complementary, cross-validating methods of measuring LD membrane behavior reveal the interplay of biophysical processes on lipid droplet monolayers.
Collapse
Affiliation(s)
- Sonali A Gandhi
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA
| | - Shahnaz Parveen
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA
| | - Munirah Alduhailan
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA
| | - Ramesh Tripathi
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA
| | - Nasser Junedi
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA
| | - Mohammad Saqallah
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA
| | - Matthew A Sanders
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 40201, USA; Center for Integrative Metabolic and Endocrine Research, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Peter M Hoffmann
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA; Physical Sciences Department, Embry-Riddle Aeronautical University, Daytona Beach, FL 32114, USA
| | - Katherine Truex
- Department of Physics, United States Naval Academy, Annapolis, MD 21402, USA
| | - James G Granneman
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI 40201, USA; Center for Integrative Metabolic and Endocrine Research, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Christopher V Kelly
- Department of Physics and Astronomy, Wayne State University, Detroit, MI 48201, USA; Center for Integrative Metabolic and Endocrine Research, School of Medicine, Wayne State University, Detroit, MI 48201, USA.
| |
Collapse
|
9
|
Dudka W, Salo VT, Mahamid J. Zooming into lipid droplet biology through the lens of electron microscopy. FEBS Lett 2024; 598:1127-1142. [PMID: 38726814 DOI: 10.1002/1873-3468.14899] [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: 01/26/2024] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/25/2024]
Abstract
Electron microscopy (EM), in its various flavors, has significantly contributed to our understanding of lipid droplets (LD) as central organelles in cellular metabolism. For example, EM has illuminated that LDs, in contrast to all other cellular organelles, are uniquely enclosed by a single phospholipid monolayer, revealed the architecture of LD contact sites with different organelles, and provided near-atomic resolution maps of key enzymes that regulate neutral lipid biosynthesis and LD biogenesis. In this review, we first provide a brief history of pivotal findings in LD biology unveiled through the lens of an electron microscope. We describe the main EM techniques used in the context of LD research and discuss their current capabilities and limitations, thereby providing a foundation for utilizing suitable EM methodology to address LD-related questions with sufficient level of structural preservation, detail, and resolution. Finally, we highlight examples where EM has recently been and is expected to be instrumental in expanding the frontiers of LD biology.
Collapse
Affiliation(s)
- Wioleta Dudka
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Veijo T Salo
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Julia Mahamid
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Cell Biology and Biophysics Unit, EMBL, Heidelberg, Germany
| |
Collapse
|
10
|
Sharma AK, Khandelwal R, Wolfrum C. Futile lipid cycling: from biochemistry to physiology. Nat Metab 2024; 6:808-824. [PMID: 38459186 DOI: 10.1038/s42255-024-01003-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
In the healthy state, the fat stored in our body isn't just inert. Rather, it is dynamically mobilized to maintain an adequate concentration of fatty acids (FAs) in our bloodstream. Our body tends to produce excess FAs to ensure that the FA availability is not limiting. The surplus FAs are actively re-esterified into glycerides, initiating a cycle of breakdown and resynthesis of glycerides. This cycle consumes energy without generating a new product and is commonly referred to as the 'futile lipid cycle' or the glyceride/FA cycle. Contrary to the notion that it's a wasteful process, it turns out this cycle is crucial for systemic metabolic homeostasis. It acts as a control point in intra-adipocyte and inter-organ cross-talk, a metabolic rheostat, an energy sensor and a lipid diversifying mechanism. In this Review, we discuss the metabolic regulation and physiological implications of the glyceride/FA cycle and its mechanistic underpinnings.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
11
|
Deng B, Kong W, Shen X, Han C, Zhao Z, Chen S, Zhou C, Bae-Jump V. The role of DGAT1 and DGAT2 in regulating tumor cell growth and their potential clinical implications. J Transl Med 2024; 22:290. [PMID: 38500157 PMCID: PMC10946154 DOI: 10.1186/s12967-024-05084-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 03/10/2024] [Indexed: 03/20/2024] Open
Abstract
Lipid metabolism is widely reprogrammed in tumor cells. Lipid droplet is a common organelle existing in most mammal cells, and its complex and dynamic functions in maintaining redox and metabolic balance, regulating endoplasmic reticulum stress, modulating chemoresistance, and providing essential biomolecules and ATP have been well established in tumor cells. The balance between lipid droplet formation and catabolism is critical to maintaining energy metabolism in tumor cells, while the process of energy metabolism affects various functions essential for tumor growth. The imbalance of synthesis and catabolism of fatty acids in tumor cells leads to the alteration of lipid droplet content in tumor cells. Diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2, the enzymes that catalyze the final step of triglyceride synthesis, participate in the formation of lipid droplets in tumor cells and in the regulation of cell proliferation, migration and invasion, chemoresistance, and prognosis in tumor. Several diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2 inhibitors have been developed over the past decade and have shown anti-tumor effects in preclinical tumor models and improvement of metabolism in clinical trials. In this review, we highlight key features of fatty acid metabolism and different paradigms of diacylglycerol acyltransferase 1 and diacylglycerol acyltransferase 2 activities on cell proliferation, migration, chemoresistance, and prognosis in tumor, with the hope that these scientific findings will have potential clinical implications.
Collapse
Affiliation(s)
- Boer Deng
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Weimin Kong
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Xiaochang Shen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chao Han
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
| | - Ziyi Zhao
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Shuning Chen
- Department of Gynecologic Oncology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chunxiao Zhou
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| | - Victoria Bae-Jump
- Division of Gynecologic Oncology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
12
|
Chauhan SS, Casillas AL, Vizzerra AD, Liou H, Clements AN, Flores CE, Prevost CT, Kashatus DF, Snider AJ, Snider JM, Warfel NA. PIM1 drives lipid droplet accumulation to promote proliferation and survival in prostate cancer. Oncogene 2024; 43:406-419. [PMID: 38097734 PMCID: PMC10837079 DOI: 10.1038/s41388-023-02914-0] [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: 07/18/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 02/04/2024]
Abstract
Lipid droplets (LDs) are dynamic organelles with a neutral lipid core surrounded by a phospholipid monolayer. Solid tumors exhibit LD accumulation, and it is believed that LDs promote cell survival by providing an energy source during energy deprivation. However, the precise mechanisms controlling LD accumulation and utilization in prostate cancer are not well known. Here, we show peroxisome proliferator-activated receptor α (PPARα) acts downstream of PIM1 kinase to accelerate LD accumulation and promote cell proliferation in prostate cancer. Mechanistically, PIM1 inactivates glycogen synthase kinase 3 beta (GSK3β) via serine 9 phosphorylation. GSK3β inhibition stabilizes PPARα and enhances the transcription of genes linked to peroxisomal biogenesis (PEX3 and PEX5) and LD growth (Tip47). The effects of PIM1 on LD accumulation are abrogated with GW6471, a specific inhibitor for PPARα. Notably, LD accumulation downstream of PIM1 provides a significant survival advantage for prostate cancer cells during nutrient stress, such as glucose depletion. Inhibiting PIM reduces LD accumulation in vivo alongside slow tumor growth and proliferation. Furthermore, TKO mice, lacking PIM isoforms, exhibit suppression in circulating triglycerides. Overall, our findings establish PIM1 as an important regulator of LD accumulation through GSK3β-PPARα signaling axis to promote cell proliferation and survival during nutrient stress.
Collapse
Affiliation(s)
- Shailender S Chauhan
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
| | - Andrea L Casillas
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Andres D Vizzerra
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA
| | - Hope Liou
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Amber N Clements
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Caitlyn E Flores
- Cancer Biology Graduate Program, University of Arizona, Tucson, AZ, 85721, USA
| | - Christopher T Prevost
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - David F Kashatus
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia Health System, Charlottesville, VA, 22908, USA
| | - Ashley J Snider
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Justin M Snider
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, 85721, USA
| | - Noel A Warfel
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, 85724, USA.
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ, 85724, USA.
| |
Collapse
|
13
|
Fang M, Liu X, Xu W, Wang X, Xu L, Zhao TJ, Li P, Yang H. Paxillin family proteins Hic-5 and LPXN promote lipid storage by regulating the ubiquitination degradation of CIDEC. J Biol Chem 2024; 300:105610. [PMID: 38159847 PMCID: PMC10850781 DOI: 10.1016/j.jbc.2023.105610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/16/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
Many metabolic diseases are caused by disorders of lipid homeostasis. CIDEC, a lipid droplet (LD)-associated protein, plays a critical role in controlling LD fusion and lipid storage. However, regulators of CIDEC remain largely unknown. Here, we established a homogeneous time-resolved fluorescence (HTRF)-based high-throughput screening method and identified LPXN as a positive regulatory candidate for CIDEC. LPXN and Hic-5, the members of the Paxillin family, are focal adhesion adaptor proteins that contribute to the recruitment of specific kinases and phosphatases, cofactors, and structural proteins, participating in the transduction of extracellular signals into intracellular responses. Our data showed that Hic-5 and LPXN significantly increased the protein level of CIDEC and enhanced CIDEC stability not through triacylglycerol synthesis and FAK signaling pathways. Hic-5 and LPXN reduced the ubiquitination of CIDEC and inhibited its proteasome degradation pathway. Furthermore, Hic-5 and LPXN enlarged LDs and promoted lipid storage in adipocytes. Therefore, we identified Hic-5 and LPXN as novel regulators of CIDEC. Our current findings also suggest intervention with Hic-5 and LPXN might ameliorate ectopic fat storage by enhancing the lipid storage capacity of white adipose tissues.
Collapse
Affiliation(s)
- Mingyu Fang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Xu Liu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Wenbo Xu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Xing Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Lin Xu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China
| | - Tong-Jin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Peng Li
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China; Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, Henan, China; Shanghai Qi Zhi Institute, Shanghai, China
| | - Hui Yang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, China.
| |
Collapse
|
14
|
Bu SY. Role of Dgat2 in Glucose Uptake and Fatty Acid Metabolism in C2C12 Skeletal Myotubes. J Microbiol Biotechnol 2023; 33:1563-1575. [PMID: 37644753 PMCID: PMC10772559 DOI: 10.4014/jmb.2307.07018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 07/25/2023] [Accepted: 07/31/2023] [Indexed: 08/31/2023]
Abstract
Acyl-coenzyme A (CoA):diacylglycerol acyltransferase 2 (DGAT2) catalyzes the last stage of triacylglycerol (TAG) synthesis, a process that forms ester bonds with diacylglycerols (DAG) and fatty acyl-CoA substrates. The enzymatic role of Dgat2 has been studied in various biological species. Still, the full description of how Dgat2 channels fatty acids in skeletal myocytes and the consequence thereof in glucose uptake have yet to be well established. Therefore, this study explored the mediating role of Dgat2 in glucose uptake and fatty acid partitioning under short interfering ribonucleic acid (siRNA)-mediated Dgat2 knockdown conditions. Cells transfected with Dgat2 siRNA downregulated glucose transporter type 4 (Glut4) messenger RNA (mRNA) expression and decreased the cellular uptake of [1-14C]-labeled 2-deoxyglucose up to 24.3% (p < 0.05). Suppression of Dgat2 deteriorated insulininduced Akt phosphorylation. Dgat2 siRNA reduced [1-14C]-labeled oleic acid incorporation into TAG, but increased the level of [1-14C]-labeled free fatty acids at 3 h after initial fatty acid loading. In an experiment of chasing radioisotope-labeled fatty acids, Dgat2 suppression augmented the level of cellular free fatty acids. It decreased the level of re-esterification of free fatty acids to TAG by 67.6% during the chase period, and the remaining pulses of phospholipids and cholesteryl esters were decreased by 34.5% and 61%, respectively. Incorporating labeled fatty acids into beta-oxidation products increased in Dgat2 siRNA transfected cells without gene expression involving fatty acid oxidation. These results indicate that Dgat2 has regulatory function in glucose uptake, possibly through the reaction of TAG with endogenously released or recycled fatty acids.
Collapse
Affiliation(s)
- So Young Bu
- Department of Food and Nutrition, College of Engineering, Daegu University, Gyeongsan 38453, Republic of Korea
| |
Collapse
|
15
|
Tang H, Zhang H, Liu D, Li S, Wang Z, Yu D, Guo ZB, Hou S, Zhou Z. Changes in physical architecture and lipids compounds in skeletal muscle from Pekin duck and Liancheng white duck. Poult Sci 2023; 102:103106. [PMID: 37837677 PMCID: PMC10585346 DOI: 10.1016/j.psj.2023.103106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/13/2023] [Accepted: 09/07/2023] [Indexed: 10/16/2023] Open
Abstract
As a complex food, meat displays various biochemical properties that are determined to a great extent by physical architecture and lipid metabolites. Pekin duck and Liancheng white duck are elite breeds with distinct characteristics. Here, we explored the development of the muscle fibers from embryonic stage to 10-wk after birth, and muscle fibers grow slowly after 8-wk. We investigated the meat quality, ultrastructure, lipidomics profiling, and lipids spatial distribution of skeletal muscle at 8 wk. Pekin duck has lower Warner-Bratzler shear force (WBSF) (P < 0.05), high intramuscular fat (IMF) (P < 0.01), longer and wider sarcomere, and higher mitochondrial density (P < 0.001). Liancheng white duck with tighter collagen architecture. A total of 950 lipids from 6 lipid classes identified with lipidomics were analyzed, the levels of GP, GL, and PR were significantly higher in Pekin duck (P < 0.05), SL and ST were significantly higher in Liancheng white duck (P < 0.05). There were 333 significantly different lipids (|log2(Fold Change)| ≥ 1 and FDR < 0.05) screened, most lipids distributed in the muscle tissue were uniform, but some specifically distributed in connective tissue. To some extent, the results demonstrate the high lipid deposition capacity of Pekin duck and the high medicinal function of Liancheng white duck. Our study provides new insights into the relationship between skeletal muscle architecture and meat toughness, which increased the knowledge of lipidomic characteristics and provide a basis for duck meat authentication.
Collapse
Affiliation(s)
- Hehe Tang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - He Zhang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Dapeng Liu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Shunan Li
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Zhen Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Daxun Yu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Zhan Bao Guo
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Shuisheng Hou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Zhengkui Zhou
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China.
| |
Collapse
|
16
|
Hou X, Zhang R, Yang M, Niu N, Zong W, Yang L, Li H, Hou R, Wang X, Wang L, Liu X, Shi L, Zhao F, Wang L, Zhang L. Characteristics of Transcriptome and Metabolome Concerning Intramuscular Fat Content in Beijing Black Pigs. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:15874-15883. [PMID: 37847170 DOI: 10.1021/acs.jafc.3c02669] [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: 10/18/2023]
Abstract
To study the characteristics of genes and metabolites related to intramuscular fat (IMF) content with less influence by breed background and individual differences, the skeletal muscle samples from 40 Beijing black pigs with either high or low IMF content were used to perform transcriptome and metabolome analyses. About 99 genes (twofold-change) were differentially expressed. Up-regulated genes in the high IMF pigs were mainly related to fat metabolism. The key genes in charge of IMF deposition are ADIPOQ, CIDEC, CYP4B1, DGAT2, LEP, OPRL1, PLIN1, SCD, and THRSP. KLHL40, TRAFD1, and HSPA6 were novel candidate genes for the IMF trait due to their high abundances. In the low IMF pigs, the differentially expressed genes involved in virus resistance were up-regulated. About 16 and 18 differential metabolites (1.5 fold-change) were obtained in the positive and negative modes, respectively. Pigs with low IMF had weaker fatty acid oxidation due to the down-regulation of various carnitines. Differentially expressed genes were more important in determining IMF deposition than differential metabolites because relatively few differential metabolites were obtained, and they were merely the products under the physiological status of diverged IMF content. This study provided valuable information for further studies on IMF deposition.
Collapse
Affiliation(s)
- Xinhua Hou
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Run Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Man Yang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Naiqi Niu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Wencheng Zong
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Liyu Yang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Huihui Li
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Renda Hou
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Xiaoqing Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Ligang Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Xin Liu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Lijun Shi
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Fuping Zhao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Lixian Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| | - Longchao Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, PR China
| |
Collapse
|
17
|
Prado LG, Camara NOS, Barbosa AS. Cell lipid biology in infections: an overview. Front Cell Infect Microbiol 2023; 13:1148383. [PMID: 37868347 PMCID: PMC10587689 DOI: 10.3389/fcimb.2023.1148383] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 09/22/2023] [Indexed: 10/24/2023] Open
Abstract
Lipids are a big family of molecules with a vast number of functions in the cell membranes, within the cytoplasm, and extracellularly. Lipid droplets (LDs) are the most common storage organelles and are present in almost every tissue type in the body. They also have structural functions serving as building blocks of cellular membranes and may be precursors of other molecules such as hormones, and lipoproteins, and as messengers in signal transduction. Fatty acids (FAs), such as sterol esters and triacylglycerols, are stored in LDs and are used in β-oxidation as fuel for tricarboxylic acid cycle (TCA) and adenosine triphosphate (ATP) generation. FA uptake and entrance in the cytoplasm are mediated by membrane receptors. After a cytoplasmic round of α- and β-oxidation, FAs are guided into the mitochondrial matrix by the L-carnitine shuttle system, where they are fully metabolized, and enter the TCA cycle. Pathogen infections may lead to impaired lipid metabolism, usage of membrane phospholipids, and LD accumulation in the cytoplasm of infected cells. Otherwise, bacterial pathogens may use lipid metabolism as a carbon source, thus altering the reactions and leading to cellular and organelles malfunctioning. This review aims to describe cellular lipid metabolism and alterations that occur upon infections.
Collapse
Affiliation(s)
- Luan Gavião Prado
- Laboratório de Bacteriologia, Instituto Butantan, São Paulo, Brazil
- Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Niels Olsen Saraiva Camara
- Laboratório de Imunobiologia de Transplantes, Departamento de Imunologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
18
|
Abstract
The sorting and trafficking of lipids between organelles gives rise to a dichotomy of bulk membrane properties between organelles of the secretory and endolysosome networks, giving rise to two "membrane territories" based on differences in lipid-packing density, net membrane charge, and bilayer leaflet asymmetries. The cellular organelle membrane dichotomy emerges from ER-to-PM anterograde membrane trafficking and the synthesis of sphingolipids and cholesterol flux at the trans-Golgi network, which constitutes the interface between the two membrane territories. Organelle homeostasis is maintained by vesicle-mediated retrieval of bulk membrane from the distal organelles of each territory to the endoplasmic reticulum or plasma membrane and by soluble lipid transfer proteins that traffic particular lipids. The concept of cellular membrane territories emphasizes the contrasting features of organelle membranes of the secretory and endolysosome networks and the essential roles of lipid-sorting pathways that maintain organelle function.
Collapse
Affiliation(s)
- Yeongho Kim
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | - Christopher G Burd
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| |
Collapse
|
19
|
Xie J, Hou X, He W, Xiao J, Cao Y, Liu X. Astaxanthin reduces fat storage in a fat-6/ fat-7 dependent manner determined using high fat Caenorhabditis elegans. Food Funct 2023; 14:7347-7360. [PMID: 37490309 DOI: 10.1039/d3fo01403g] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Although astaxanthin has been shown to have high potential for weight loss, the specific action site and signal pathway generally cannot be confirmed in other animal models. This prevents us from finding therapeutic targets. Hence, we further illuminated its efficacy and specific action sites by using Caenorhabditis elegans (C. elegans). In this study, 60 μM astaxanthin supplementation reduced overall fat deposition and triglyceride levels by 21.47% and 22.00% (p < 0.01). The content of large lipid droplets was reversed after astaxanthin treatment, and the ratio of oleic acid/stearic acid (C18:1Δ9/C18:0) decreased significantly, which were essential substrates for triglyceride biosynthesis. In addition, astaxanthin prevented obesity caused by excessive energy accumulation and insufficient energy consumption. Furthermore, the above effects were induced by sbp-1/mdt-15 and insulin/insulin-like growth factor pathways, and finally co-regulated the specific site-fat-6 and fat-7 down-regulation. These results provided insight into therapeutic targets for future astaxanthin as a nutritional health product to relieve obesity.
Collapse
Affiliation(s)
- Junting Xie
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Xiaoning Hou
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Wanshi He
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Xiaojuan Liu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou, 510642, Guangdong, China.
- Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| |
Collapse
|
20
|
Gandhi SA, Parveen S, Alduhailan M, Tripathi R, Junedi N, Saqallah M, Sanders MA, Hoffmann PM, Truex K, Granneman JG, Kelly CV. Methods for making and observing model lipid droplets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.17.549385. [PMID: 37503132 PMCID: PMC10370146 DOI: 10.1101/2023.07.17.549385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The mechanisms by which the lipid droplet (LD) membrane is remodeled in concert with the activation of lipolysis incorporate a complex interplay of proteins, phospholipids, and neutral lipids. Model LDs (mLDs) provide an isolated, purified system for testing the mechanisms by which the droplet composition, size, shape, and tension affects triglyceride metabolism. Described here are methods of making and testing mLDs ranging from 0.1 to 40 μm diameter with known composition. Methods are described for imaging mLDs with high-resolution microscopy during buffer exchanges for the measurement of membrane binding, diffusion, and tension via fluorescence correlation spectroscopy (FCS), fluorescence recovery after photobleaching (FRAP), fluorescence lifetime imaging microscopy (FLIM), atomic force microscopy (AFM), pendant droplet tensiometry, and imaging flow cytometry. These complementary, cross-validating methods of measuring LD membrane behavior reveal the interplay of biophysical processes in triglyceride metabolism.
Collapse
Affiliation(s)
- Sonali A. Gandhi
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
| | - Shahnaz Parveen
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
| | - Munirah Alduhailan
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
| | - Ramesh Tripathi
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
| | - Nasser Junedi
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
| | - Mohammad Saqallah
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
| | - Matthew A. Sanders
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, USA 40201
- Center for Integrative Metabolic and Endocrine Research, School of Medicine, Wayne State University, Detroit, MI USA 48201
| | - Peter M. Hoffmann
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
- Physical Sciences Department, Embry-Riddle Aeronautical University, Daytona Beach, FL, USA 32114
| | - Katherine Truex
- Department of Physics, United States Naval Academy, Annapolis, MD, USA 21402
| | - James G. Granneman
- Center for Molecular Medicine and Genetics, School of Medicine, Wayne State University, Detroit, MI, USA 40201
- Center for Integrative Metabolic and Endocrine Research, School of Medicine, Wayne State University, Detroit, MI USA 48201
| | - Christopher V Kelly
- Department of Physics and Astronomy, Wayne State University, Detroit, MI, USA 48201
- Center for Integrative Metabolic and Endocrine Research, School of Medicine, Wayne State University, Detroit, MI USA 48201
| |
Collapse
|
21
|
Hammoudeh N, Soukkarieh C, Murphy DJ, Hanano A. Mammalian lipid droplets: structural, pathological, immunological and anti-toxicological roles. Prog Lipid Res 2023; 91:101233. [PMID: 37156444 DOI: 10.1016/j.plipres.2023.101233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/30/2023] [Accepted: 05/05/2023] [Indexed: 05/10/2023]
Abstract
Mammalian lipid droplets (LDs) are specialized cytosolic organelles consisting of a neutral lipid core surrounded by a membrane made up of a phospholipid monolayer and a specific population of proteins that varies according to the location and function of each LD. Over the past decade, there have been significant advances in the understanding of LD biogenesis and functions. LDs are now recognized as dynamic organelles that participate in many aspects of cellular homeostasis plus other vital functions. LD biogenesis is a complex, highly-regulated process with assembly occurring on the endoplasmic reticulum although aspects of the underpinning molecular mechanisms remain elusive. For example, it is unclear how many enzymes participate in the biosynthesis of the neutral lipid components of LDs and how this process is coordinated in response to different metabolic cues to promote or suppress LD formation and turnover. In addition to enzymes involved in the biosynthesis of neutral lipids, various scaffolding proteins play roles in coordinating LD formation. Despite their lack of ultrastructural diversity, LDs in different mammalian cell types are involved in a wide range of biological functions. These include roles in membrane homeostasis, regulation of hypoxia, neoplastic inflammatory responses, cellular oxidative status, lipid peroxidation, and protection against potentially toxic intracellular fatty acids and lipophilic xenobiotics. Herein, the roles of mammalian LDs and their associated proteins are reviewed with a particular focus on their roles in pathological, immunological and anti-toxicological processes.
Collapse
Affiliation(s)
- Nour Hammoudeh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Chadi Soukkarieh
- Department of Animal Biology, Faculty of Sciences, University of Damascus, Damascus, Syria
| | - Denis J Murphy
- School of Applied Sciences, University of South Wales, Pontypridd, CF37 1DL, Wales, United Kingdom..
| | - Abdulsamie Hanano
- Department of Molecular Biology and Biotechnology, Atomic Energy Commission of Syria (AECS), P.O. Box 6091, Damascus, Syria..
| |
Collapse
|
22
|
Walther TC, Kim S, Arlt H, Voth GA, Farese RV. Structure and function of lipid droplet assembly complexes. Curr Opin Struct Biol 2023; 80:102606. [PMID: 37150040 PMCID: PMC10853036 DOI: 10.1016/j.sbi.2023.102606] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/03/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023]
Abstract
Cells store lipids as a reservoir of metabolic energy and membrane component precursors in organelles called lipid droplets (LDs). LD formation occurs in the endoplasmic reticulum (ER) at LD assembly complexes (LDAC), consisting of an oligomeric core of seipin and accessory proteins. LDACs determine the sites of LD formation and are required for this process to occur normally. Seipin oligomers form a cage-like structure in the membrane that may serve to facilitate the phase transition of neutral lipids in the membrane to form an oil droplet within the LDAC. Modeling suggests that, as the LD grows, seipin anchors it to the ER bilayer and conformational shifts of seipin transmembrane segments open the LDAC dome toward the cytoplasm, enabling the emerging LD to egress from the ER.
Collapse
Affiliation(s)
- Tobias C Walther
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Howard Hughes Medical Institute, New York, NY 10065, USA.
| | - Siyoung Kim
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, 60637, USA
| | - Henning Arlt
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Gregory A Voth
- Department of Chemistry, Chicago Center for Theoretical Chemistry, James Franck Institute, and Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, 60637, USA
| | - Robert V Farese
- Cell Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| |
Collapse
|
23
|
Farese RV, Walther TC. Glycerolipid Synthesis and Lipid Droplet Formation in the Endoplasmic Reticulum. Cold Spring Harb Perspect Biol 2023; 15:a041246. [PMID: 36096640 PMCID: PMC10153804 DOI: 10.1101/cshperspect.a041246] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
More than 60 years ago, Eugene Kennedy and coworkers elucidated the endoplasmic reticulum (ER)-based pathways of glycerolipid synthesis, including the synthesis of phospholipids and triacylglycerols (TGs). The reactions of the Kennedy pathway were identified by studying the conversion of lipid intermediates and the isolation of biochemical enzymatic activities, but the molecular basis for most of these reactions was unknown. With recent progress in the cell biology, biochemistry, and structural biology in this area, we have a much more mechanistic understanding of this pathway and its reactions. In this review, we provide an overview of molecular aspects of glycerolipid synthesis, focusing on recent insights into the synthesis of TGs. Further, we go beyond the Kennedy pathway to describe the mechanisms for storage of TG in cytosolic lipid droplets and discuss how overwhelming these pathways leads to ER stress and cellular toxicity, as seen in diseases linked to lipid overload and obesity.
Collapse
Affiliation(s)
- Robert V Farese
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Center for Causes and Prevention of Cardiovascular Disease (CAP-CVD), Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Tobias C Walther
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts 02142, USA
- Center for Causes and Prevention of Cardiovascular Disease (CAP-CVD), Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
- Howard Hughes Medical Institute Boston, Boston, Massachusetts 02115, USA
| |
Collapse
|
24
|
Ratnayake OC, Chotiwan N, Saavedra-Rodriguez K, Perera R. The buzz in the field: the interaction between viruses, mosquitoes, and metabolism. Front Cell Infect Microbiol 2023; 13:1128577. [PMID: 37360524 PMCID: PMC10289420 DOI: 10.3389/fcimb.2023.1128577] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 06/28/2023] Open
Abstract
Among many medically important pathogens, arboviruses like dengue, Zika and chikungunya cause severe health and economic burdens especially in developing countries. These viruses are primarily vectored by mosquitoes. Having surmounted geographical barriers and threat of control strategies, these vectors continue to conquer many areas of the globe exposing more than half of the world's population to these viruses. Unfortunately, no medical interventions have been capable so far to produce successful vaccines or antivirals against many of these viruses. Thus, vector control remains the fundamental strategy to prevent disease transmission. The long-established understanding regarding the replication of these viruses is that they reshape both human and mosquito host cellular membranes upon infection for their replicative benefit. This leads to or is a result of significant alterations in lipid metabolism. Metabolism involves complex chemical reactions in the body that are essential for general physiological functions and survival of an organism. Finely tuned metabolic homeostases are maintained in healthy organisms. However, a simple stimulus like a viral infection can alter this homeostatic landscape driving considerable phenotypic change. Better comprehension of these mechanisms can serve as innovative control strategies against these vectors and viruses. Here, we review the metabolic basis of fundamental mosquito biology and virus-vector interactions. The cited work provides compelling evidence that targeting metabolism can be a paradigm shift and provide potent tools for vector control as well as tools to answer many unresolved questions and gaps in the field of arbovirology.
Collapse
Affiliation(s)
- Oshani C. Ratnayake
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Nunya Chotiwan
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Karla Saavedra-Rodriguez
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| | - Rushika Perera
- Center for Vector-borne Infectious Diseases, Dept. of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
25
|
Dias SSG, Cunha-Fernandes T, Souza-Moreira L, Soares VC, Lima GB, Azevedo-Quintanilha IG, Santos J, Pereira-Dutra F, Freitas C, Reis PA, Rehen SK, Bozza FA, Souza TML, de Almeida CJG, Bozza PT. Metabolic reprogramming and lipid droplets are involved in Zika virus replication in neural cells. J Neuroinflammation 2023; 20:61. [PMID: 36882750 PMCID: PMC9992922 DOI: 10.1186/s12974-023-02736-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Abstract
Zika virus (ZIKV) infection is a global public health concern linked to adult neurological disorders and congenital diseases in newborns. Host lipid metabolism, including lipid droplet (LD) biogenesis, has been associated with viral replication and pathogenesis of different viruses. However, the mechanisms of LD formation and their roles in ZIKV infection in neural cells are still unclear. Here, we demonstrate that ZIKV regulates the expression of pathways associated with lipid metabolism, including the upregulation and activation of lipogenesis-associated transcription factors and decreased expression of lipolysis-associated proteins, leading to significant LD accumulation in human neuroblastoma SH-SY5Y cells and in neural stem cells (NSCs). Pharmacological inhibition of DGAT-1 decreased LD accumulation and ZIKV replication in vitro in human cells and in an in vivo mouse model of infection. In accordance with the role of LDs in the regulation of inflammation and innate immunity, we show that blocking LD formation has major roles in inflammatory cytokine production in the brain. Moreover, we observed that inhibition of DGAT-1 inhibited the weight loss and mortality induced by ZIKV infection in vivo. Our results reveal that LD biogenesis triggered by ZIKV infection is a crucial step for ZIKV replication and pathogenesis in neural cells. Therefore, targeting lipid metabolism and LD biogenesis may represent potential strategies for anti-ZIKV treatment development.
Collapse
Affiliation(s)
- Suelen Silva Gomes Dias
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Tamires Cunha-Fernandes
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Luciana Souza-Moreira
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Vinicius Cardoso Soares
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.,Programa de Imunologia e Inflamação, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giselle Barbosa Lima
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | | | - Julia Santos
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Filipe Pereira-Dutra
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Caroline Freitas
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Patricia A Reis
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.,Departamento de Bioquímica, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Stevens Kastrup Rehen
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, Brazil.,Instituto de Biologia, Universidade Federal do Rio de Janeiro, UFRJ, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando A Bozza
- Instituto D'Or de Pesquisa e Ensino (IDOR), Rio de Janeiro, Brazil.,Instituto Nacional de Infectologia Evandro Chagas (INI), FIOCRUZ, Rio de Janeiro, Brazil
| | - Thiago M Lopes Souza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.,Instituto Nacional de Ciência e Tecnologia em Inovação em Doenças de Populações Negligenciadas (INCT/IDPN), Centro de Desenvolvimento Tecnológico em Saúde, (CDTS), FIOCRUZ, Rio de Janeiro, Brazil
| | - Cecilia J G de Almeida
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil
| | - Patricia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (FIOCRUZ), Rio de Janeiro, Brazil.
| |
Collapse
|
26
|
Livelo C, Guo Y, Abou Daya F, Rajasekaran V, Varshney S, Le HD, Barnes S, Panda S, Melkani GC. Time-restricted feeding promotes muscle function through purine cycle and AMPK signaling in Drosophila obesity models. Nat Commun 2023; 14:949. [PMID: 36810287 PMCID: PMC9944249 DOI: 10.1038/s41467-023-36474-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 02/01/2023] [Indexed: 02/23/2023] Open
Abstract
Obesity caused by genetic and environmental factors can lead to compromised skeletal muscle function. Time-restricted feeding (TRF) has been shown to prevent muscle function decline from obesogenic challenges; however, its mechanism remains unclear. Here we demonstrate that TRF upregulates genes involved in glycine production (Sardh and CG5955) and utilization (Gnmt), while Dgat2, involved in triglyceride synthesis is downregulated in Drosophila models of diet- and genetic-induced obesity. Muscle-specific knockdown of Gnmt, Sardh, and CG5955 lead to muscle dysfunction, ectopic lipid accumulation, and loss of TRF-mediated benefits, while knockdown of Dgat2 retains muscle function during aging and reduces ectopic lipid accumulation. Further analyses demonstrate that TRF upregulates the purine cycle in a diet-induced obesity model and AMPK signaling-associated pathways in a genetic-induced obesity model. Overall, our data suggest that TRF improves muscle function through modulations of common and distinct pathways under different obesogenic challenges and provides potential targets for obesity treatments.
Collapse
Affiliation(s)
- Christopher Livelo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Yiming Guo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Farah Abou Daya
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Vasanthi Rajasekaran
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Shweta Varshney
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Department of Biology, Molecular Biology Institute, San Diego State University, San Diego, CA, 92182, USA
| | - Hiep D Le
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Stephen Barnes
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Girish C Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
- Department of Biology, Molecular Biology Institute, San Diego State University, San Diego, CA, 92182, USA.
| |
Collapse
|
27
|
Multiscale spectroscopic analysis of lipids in dimorphic and oleaginous Mucor circinelloides accommodate sustainable targeted lipid production. Fungal Biol Biotechnol 2023; 10:2. [PMID: 36647105 PMCID: PMC9843973 DOI: 10.1186/s40694-023-00148-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 01/05/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Oleaginous fungi have versatile metabolism and able to transform a wide range of substrates into lipids, accounting up to 20-70% of their total cell mass. Therefore, oleaginous fungi are considered as an alternative source of lipids. Oleaginous fungi can accumulate mainly acyl glycerides and free fatty acids which are localized in lipid droplets. Some of the oleaginous fungi possessing promising lipid productivity are dimorphic and can exhibit three cell forms, flat hyphae, swollen hyphae and yeast-like cells. To develop sustainable targeted fungal lipid production, deep understanding of lipogenesis and lipid droplet chemistry in these cell forms is needed at multiscale level. In this study, we explored the potential of infrared spectroscopy techniques for examining lipid droplet formation and accumulation in different cell forms of the dimorphic and oleaginous fungus Mucor circinelloides. RESULTS Both transmission- and reflectance-based spectroscopy techniques are shown to be well suited for studying bulk fungal biomass. Exploring single cells with infrared microspectroscopy reveals differences in chemical profiles and, consequently, lipogenesis process, for different cell forms. Yeast-like cells of M. circinelloides exhibited the highest absorbance intensities for lipid-associated peaks in comparison to hyphae-like cell forms. Lipid-to-protein ratio, which is commonly used in IR spectroscopy to estimate lipid yield was the lowest in flat hyphae. Swollen hyphae are mainly composed of lipids and characterized by more uniform distribution of lipid-to-protein concentration. Yeast-like cells seem to be comprised mostly of lipids having the largest lipid-to-protein ratio among all studied cell forms. With infrared nanospectroscopy, variations in the ratios between lipid fractions triglycerides and free fatty acids and clear evidence of heterogeneity within and between lipid droplets are illustrated for the first time. CONCLUSIONS Vibrational spectroscopy techniques can provide comprehensive information on lipogenesis in dimorphic and oleaginous fungi at the levels of the bulk of cells, single cells and single lipid droplets. Unicellular spectra showed that various cell forms of M. circinelloides differs in the total lipid content and profile of the accumulated lipids, where yeast-like cells are the fatty ones and, therefore, could be considered as preferable cell form for producing lipid-rich biomass. Spectra of single lipid droplets showed an indication of possible droplet-to-droplet and within-droplet heterogeneity.
Collapse
|
28
|
Li P, Tian Y, Du M, Xie Q, Chen Y, Ma L, Huang Y, Yin Z, Xu H, Wu X. Mechanism of Rotenone Toxicity against Plutella xylostella: New Perspective from a Spatial Metabolomics and Lipidomics Study. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:211-222. [PMID: 36538414 DOI: 10.1021/acs.jafc.2c06292] [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: 06/17/2023]
Abstract
The botanical pesticide rotenone can effectively control target pest Plutella xylostella, yet insights into in situ metabolic regulation of P. xylostella toward rotenone remain limited. Herein, we demonstrated metabolic expression levels and spatial distribution of rotenone-treated P. xylostella using spatial metabolomics and lipidomics. Specifically, rotenone significantly affected purine and amino acid metabolisms, indicating that adenosine monophosphate and inosine were distributed in the whole body of P. xylostella with elevated levels, while guanosine 5'-monophosphate and tryptophan were significantly downregulated. Spatial lipidomics results indicated that rotenone may significantly destroy glycerophospholipids in cell membranes of P. xylostella, inhibit fatty acid biosynthesis, and consume diacylglycerol to enhance fat oxidation. These findings revealed that high toxicity of rotenone toward P. xylostella may be ascribed to negative effects on energy production and amino acid synthesis and damage to cell membranes, providing guidelines for the toxicity mechanism of rotenone on target pests and rational development of botanical pesticide candidates.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
- Key Laboratory of Bio-Pesticide Creation and Application of Guangdong Province, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
| | - Yongqing Tian
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Mingyi Du
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Qingrong Xie
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Yingying Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Lianlian Ma
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Yudi Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Zhibin Yin
- Agro-biological Gene Research Center, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Hanhong Xu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| | - Xinzhou Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources and Key Laboratory of Natural Pesticide and Chemical Biology of the Ministry of Education, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
29
|
Amin NB, Saxena AR, Somayaji V, Dullea R. Inhibition of Diacylglycerol Acyltransferase 2 Versus Diacylglycerol Acyltransferase 1: Potential Therapeutic Implications of Pharmacology. Clin Ther 2023; 45:55-70. [PMID: 36690550 DOI: 10.1016/j.clinthera.2022.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 12/01/2022] [Accepted: 12/15/2022] [Indexed: 01/22/2023]
Abstract
PURPOSE Hepatic steatosis due to altered lipid metabolism and accumulation of hepatic triglycerides is a hallmark of nonalcoholic fatty liver disease (NAFLD). Diacylglycerol acyltransferase (DGAT) enzymes, DGAT1 and DGAT2, catalyze the terminal reaction in triglyceride synthesis, making them attractive targets for pharmacologic intervention. There is a common misconception that these enzymes are related; however, despite their similar names, DGAT1 and DGAT2 differ significantly on multiple levels. As we look ahead to future clinical studies of DGAT2 inhibitors in patients with NAFLD and nonalcoholic steatohepatitis (NASH), we review key differences and include evidence to highlight and support DGAT2 inhibitor (DGAT2i) pharmacology. METHODS Three Phase I, randomized, double-blind, placebo-controlled trials assessed the safety, tolerability, and pharmacokinetic properties of the DGAT2i ervogastat (PF-06865571) in healthy adult participants (Single Dose Study to Assess the Safety, Tolerability and Pharmacokinetics of PF-06865571 [study C2541001] and Study to Assess the Safety, Tolerability, and Pharmacokinetics of Multiple Doses of PF-06865571 in Healthy, Including Overweight and Obese, Adult Subjects [study C2541002]) or participants with NAFLD (2-Week Study in People With Nonalcoholic Fatty Liver Disease [study C2541005]). Data from 2 Phase I, randomized, double-blind, placebo-controlled trials of the DGAT1i PF-04620110 in healthy participants (A Single Dose Study of PF-04620110 in Overweight and Obese, Otherwise Healthy Volunteers [study B0961001] and A Multiple Dose Study of PF-04620110 in Overweight and Obese, Otherwise Healthy Volunteers [study B0961002]) were included for comparison. Safety outcomes were the primary end point in all studies, except in study C2541005, in which safety was the secondary end point, with relative change from baseline in whole liver fat at day 15 assessed as the primary end point. Safety data were analyzed across studies by total daily dose of ervogastat (5, 15, 50, 100, 150, 500, 600, 1000, and 1500 mg) or PF-04620110 (0.3, 1, 3, 5, 7, 10, 14, and 21 mg), with placebo data pooled separately across ervogastat and PF-04620110 studies. FINDINGS Published data indicate that DGAT1 and DGAT2 differ in multiple dimensions, including gene family, subcellular localization, substrate preference, and specificity, with unrelated pharmacologic inhibition properties and differing safety profiles. Although initial nonclinical studies suggested a potentially attractive therapeutic profile with DGAT1 inhibition, genetic and pharmacologic data suggest otherwise, with common gastrointestinal adverse events, including nausea, vomiting, and diarrhea, limiting further clinical development. Conversely, DGAT2 inhibition, although initially not pursued as aggressively as a potential target for pharmacologic intervention, has consistent efficacy in nonclinical studies, with reduced triglyceride synthesis accompanied by reduced expression of genes essential for de novo lipogenesis. In addition, early clinical data indicate antisteatotic effects with DGAT2i ervogastat, in participants with NAFLD, accompanied by a well-tolerated safety profile. IMPLICATIONS Although pharmacologic DGAT1is are limited by an adverse safety profile, data support use of DGAT2i as an effective and well-tolerated therapeutic strategy for patients with NAFLD, NASH, and NASH with liver fibrosis. CLINICALTRIALS gov identifiers: NCT03092232, NCT03230383, NCT03513588, NCT00799006, and NCT00959426.
Collapse
Affiliation(s)
- Neeta B Amin
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Aditi R Saxena
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts
| | - Veena Somayaji
- Early Clinical Development, Pfizer Inc, Cambridge, Massachusetts
| | - Robert Dullea
- Internal Medicine Research Unit, Pfizer Inc, Cambridge, Massachusetts.
| |
Collapse
|
30
|
Parchuri P, Pappanoor A, Naeem A, Durrett TP, Welti R, R V S. Lipidome analysis and characterization of Buglossoides arvensis acyltransferases that incorporate polyunsaturated fatty acids into triacylglycerols. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2022; 324:111445. [PMID: 36037983 DOI: 10.1016/j.plantsci.2022.111445] [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: 06/28/2022] [Revised: 07/26/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
Buglossoides arvensis is a burgeoning oilseed crop that contains an unique combination of ω-3 and ω-6 polyunsaturated fatty acids (PUFA), constituting ~80-85% of seed triacylglycerols (TAGs). To uncover the critical TAG biosynthetic pathways contributing for high PUFA accumulation, we performed lipidome of developing seeds and characterized acyltransferases involved in the final step of TAG biosynthesis. During seed development, distribution of lipid molecular species in individual lipid classes showed distinct patterns from an early-stage (6 days after flowering (DAF)) to the middle-stage (12 and 18 DAF) of oil biosynthesis. PUFA-containing TAG species drastically increased from 6 to 12 DAF. The expression profiles of key triacylglycerol biosynthesis genes and patterns of phosphatidylcholine, diacylglycerol and triacylglycerol molecular species during seed development were used to predict the contribution of diacylglycerol acyltransferases (DGAT1 and DGAT2) and phospholipid: diacylglycerol acyltransferases (PDAT1 and PDAT2) to PUFA-rich TAG biosynthesis. Our analysis suggests that DGATs play a crucial role in enriching TAGs with PUFA compared to PDATs. This was further confirmed by fatty acid feeding studies in yeast expressing acyltransferases. BaDGAT2 preferentially incorporated high amounts of PUFAs into TAG, compared to BaDGAT1. Our results provide insight into the molecular mechanisms of TAG accumulation in this plant and identify target genes for transgenic production of SDA in traditional oilseed crops.
Collapse
Affiliation(s)
- Prasad Parchuri
- Plant Cell Biotechnology Department, CSIR-Central Food Technological Research Institute (CSIR-CFTRI), Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India; Kansas Lipidomics Research Center, Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | - Anjali Pappanoor
- Plant Cell Biotechnology Department, CSIR-Central Food Technological Research Institute (CSIR-CFTRI), Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| | - Abdulrahman Naeem
- Kansas Lipidomics Research Center, Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | - Timothy P Durrett
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS 66506, USA.
| | - Ruth Welti
- Kansas Lipidomics Research Center, Division of Biology, Kansas State University, Manhattan, KS 66506, USA.
| | - Sreedhar R V
- Plant Cell Biotechnology Department, CSIR-Central Food Technological Research Institute (CSIR-CFTRI), Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
31
|
Okotrub SV, Lebedeva DA, Okotrub KA, Chuyko EA, Brusentsev EY, Rakhmanova TA, Amstislavsky SY. Effects of Linoleic Acid on Cryopreservation of IVF-Obtained Domestic Cat Embryos. Russ J Dev Biol 2022. [DOI: 10.1134/s106236042205006x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
32
|
He Y, Su J, Gao H, Li J, Feng Z, Yin Y. Untargeted Metabolomics Reveals the Function of GPRC6A in Amino Acid and Lipid Metabolism in Mice. Metabolites 2022; 12:metabo12090776. [PMID: 36144181 PMCID: PMC9502419 DOI: 10.3390/metabo12090776] [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: 08/12/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
GPRC6A is an amino acid sensor in the cytomembrane. Despite substantial evidence for the role of GPRC6A in metabolism, the specific effects and mechanism by which this gene acts on metabolic processes are still unresolved. In this study, serum biochemical parameters related to liver and kidney function and serum amino acid levels were determined in GPRC6A wild-type (WT) and knockout (KO) mice. An untargeted serum metabolomics analysis was also conducted for the first time, to the best of our knowledge, to decipher the function of GPRC6A in metabolic processes. GPRC6A was involved in lipid and amino acid metabolism, mainly by affecting liver function. A loss of GPRC6A function may perturb bile acid metabolism, thus leading to abnormal unsaturated fatty acid metabolism. GPRC6A KO may lead to excessive protein breakdown under starvation, and the loss of GPRC6A had a significant effect on phenylalanine metabolism-related pathways. Our metabolomics data provide a novel basis for further functional studies of GPRC6A.
Collapse
Affiliation(s)
- Yumin He
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Canter for Healthy Livestock and Poultry Production, Scientific Observational and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China(
- Animal Nutrition and Human Health Laboratory, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Jingyun Su
- Animal Nutrition and Human Health Laboratory, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Hongrui Gao
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Canter for Healthy Livestock and Poultry Production, Scientific Observational and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China(
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jianzhong Li
- Animal Nutrition and Human Health Laboratory, College of Life Sciences, Hunan Normal University, Changsha 410081, China
- Correspondence: (J.L.); (Z.F.)
| | - Zemeng Feng
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Canter for Healthy Livestock and Poultry Production, Scientific Observational and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China(
- Correspondence: (J.L.); (Z.F.)
| | - Yulong Yin
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Hunan Provincial Engineering Research Canter for Healthy Livestock and Poultry Production, Scientific Observational and Experimental Station of Animal Nutrition and Feed Science in South-Central, Ministry of Agriculture, Changsha 410125, China(
- Animal Nutrition and Human Health Laboratory, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| |
Collapse
|
33
|
Schwehr BJ, Hartnell D, Massi M, Hackett MJ. Luminescent Metal Complexes as Emerging Tools for Lipid Imaging. Top Curr Chem (Cham) 2022; 380:46. [PMID: 35976575 PMCID: PMC9385838 DOI: 10.1007/s41061-022-00400-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 06/20/2022] [Indexed: 12/01/2022]
Abstract
Fluorescence microscopy is a key tool in the biological sciences, which finds use as a routine laboratory technique (e.g., epifluorescence microscope) or more advanced confocal, two-photon, and super-resolution applications. Through continued developments in microscopy, and other analytical methods, the importance of lipids as constituents of subcellular organelles, signalling or regulating molecules continues to emerge. The increasing recognition of the importance of lipids to fundamental cell biology (in health and disease) has prompted the development of protocols and techniques to image the distribution of lipids in cells and tissues. A diverse suite of spectroscopic and microscopy tools are continuously being developed and explored to add to the "toolbox" to study lipid biology. A relatively recent breakthrough in this field has been the development and subsequent application of metal-based luminescent complexes for imaging lipids in biological systems. These metal-based compounds appear to offer advantages with respect to their tunability of the photophysical properties, in addition to capabilities centred around selectively targeting specific lipid structures or classes of lipids. The presence of the metal centre also opens the path to alternative imaging modalities that might not be applicable to traditional organic fluorophores. This review examines the current progress and developments in metal-based luminescent complexes to study lipids, in addition to exploring potential new avenues and challenges for the field to take.
Collapse
Affiliation(s)
- Bradley J Schwehr
- School of Molecular and Life Sciences, Curtin University, Perth, WA, 6845, Australia
| | - David Hartnell
- School of Molecular and Life Sciences, Curtin University, Perth, WA, 6845, Australia.,Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6845, Australia
| | - Massimiliano Massi
- School of Molecular and Life Sciences, Curtin University, Perth, WA, 6845, Australia.
| | - Mark J Hackett
- School of Molecular and Life Sciences, Curtin University, Perth, WA, 6845, Australia. .,Curtin Health Innovation Research Institute, Curtin University, Perth, WA, 6845, Australia.
| |
Collapse
|
34
|
Wang Y, Li C, Zhang J, Xu X, Fu L, Xu J, Zhu H, Hu Y, Li C, Wang M, Wu Y, Zou X, Liang B. Polyunsaturated fatty acids promote the rapid fusion of lipid droplets in Caenorhabditis elegans. J Biol Chem 2022; 298:102179. [PMID: 35752365 PMCID: PMC9352923 DOI: 10.1016/j.jbc.2022.102179] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 06/07/2022] [Accepted: 06/09/2022] [Indexed: 11/27/2022] Open
Abstract
Lipid droplets (LDs) are intracellular organelles that dynamically regulate lipids and energy homeostasis in the cell. LDs can grow through either local lipid synthesis or LD fusion. However, how lipids involving in LD fusion for LD growth is largely unknown. Here, we show that genetic mutation of acox-3 (acyl-CoA oxidase), maoc-1 (enoyl-CoA hydratase), dhs-28 (3-hydroxylacyl-CoA dehydrogenase), and daf-22 (3-ketoacyl-CoA thiolase), all involved in the peroxisomal β-oxidation pathway in Caenorhabditis elegans, led to rapid fusion of adjacent LDs to form giant LDs (gLDs). Mechanistically, we show that dysfunction of peroxisomal β-oxidation results in the accumulation of long-chain fatty acid-CoA and phosphocholine, which may activate the sterol-binding protein 1/sterol regulatory element-binding protein to promote gLD formation. Furthermore, we found that inactivation of either FAT-2 (delta-12 desaturase) or FAT-3 and FAT-1 (delta-15 desaturase and delta-6 desaturase, respectively) to block the biosynthesis of polyunsaturated fatty acids (PUFAs) with three or more double bonds (n≥3-PUFAs) fully repressed the formation of gLDs; in contrast, dietary supplementation of n≥3-PUFAs or phosphocholine bearing these PUFAs led to recovery of the formation of gLDs in peroxisomal β-oxidation-defective worms lacking PUFA biosynthesis. Thus, we conclude that n≥3-PUFAs, distinct from other well-known lipids and proteins, promote rapid LD fusion leading to LD growth.
Collapse
Affiliation(s)
- Yanli Wang
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Chunxia Li
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Jingjing Zhang
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Xiumei Xu
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Lin Fu
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Jie Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Kunming Institute of Zoology, CAS, Kunming, Yunnan, China
| | - Hong Zhu
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Ying Hu
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Chengbin Li
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Mengjie Wang
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China
| | - Yingjie Wu
- Shandong Provincial Hospital, School of Laboratory Animal & Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China; Institute for Genome Engineered Animal Models of Human Diseases, National Center of Genetically Engineered Animal Models for International Research, Liaoning Provence Key Lab of Genome Engineered Animal Models Dalian Medical University, Dalian, Liaoning, China.
| | - Xiaoju Zou
- College of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| | - Bin Liang
- Center for Life Sciences, School of Life Sciences, State Key Laboratory for Conservation and Utilization of Bio-Resources, Yunnan University, Kunming, Yunnan, China.
| |
Collapse
|
35
|
Chen G, Harwood JL, Lemieux MJ, Stone SJ, Weselake RJ. Acyl-CoA:diacylglycerol acyltransferase: Properties, physiological roles, metabolic engineering and intentional control. Prog Lipid Res 2022; 88:101181. [PMID: 35820474 DOI: 10.1016/j.plipres.2022.101181] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022]
Abstract
Acyl-CoA:diacylglycerol acyltransferase (DGAT, EC 2.3.1.20) catalyzes the last reaction in the acyl-CoA-dependent biosynthesis of triacylglycerol (TAG). DGAT activity resides mainly in membrane-bound DGAT1 and DGAT2 in eukaryotes and bifunctional wax ester synthase-diacylglycerol acyltransferase (WSD) in bacteria, which are all membrane-bound proteins but exhibit no sequence homology to each other. Recent studies also identified other DGAT enzymes such as the soluble DGAT3 and diacylglycerol acetyltransferase (EaDAcT), as well as enzymes with DGAT activities including defective in cuticular ridges (DCR) and steryl and phytyl ester synthases (PESs). This review comprehensively discusses research advances on DGATs in prokaryotes and eukaryotes with a focus on their biochemical properties, physiological roles, and biotechnological and therapeutic applications. The review begins with a discussion of DGAT assay methods, followed by a systematic discussion of TAG biosynthesis and the properties and physiological role of DGATs. Thereafter, the review discusses the three-dimensional structure and insights into mechanism of action of human DGAT1, and the modeled DGAT1 from Brassica napus. The review then examines metabolic engineering strategies involving manipulation of DGAT, followed by a discussion of its therapeutic applications. DGAT in relation to improvement of livestock traits is also discussed along with DGATs in various other eukaryotic organisms.
Collapse
Affiliation(s)
- Guanqun Chen
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada.
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Membrane Protein Disease Research Group, Edmonton T6G 2H7, Canada
| | - Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| | - Randall J Weselake
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada
| |
Collapse
|
36
|
Dutta T, Pal K, Koner AL. Intracellular Physical Properties with Small Organic Fluorescent Probes: Recent Advances and Future Perspectives. CHEM REC 2022; 22:e202200035. [PMID: 35801859 DOI: 10.1002/tcr.202200035] [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] [Received: 02/21/2022] [Revised: 06/22/2022] [Indexed: 11/09/2022]
Abstract
The intracellular physical parameters i. e., polarity, viscosity, fluidity, tension, potential, and temperature of a live cell are the hallmark of cellular health and have garnered immense interest over the past decade. In this context, small molecule organic fluorophores exhibit prominent useful properties including easy functionalizability, environmental sensitivity, biocompatibility, and fast yet efficient cellular uptakability which has made them a popular tool to understand intra-cellular micro-environmental properties. Throughout this discussion, we have outlined the basic design strategies of small molecules for specific organelle targeting and quantification of physical properties. The values of these parameters are indicative of cellular homeostasis and subtle alteration may be considered as the onset of disease. We believe this comprehensive review will facilitate the development of potential future probes for superior insight into the physical parameters that are yet to be quantified.
Collapse
Affiliation(s)
- Tanoy Dutta
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, INDIA (TD) (ALK
| | - Kaushik Pal
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, INDIA (TD) (ALK.,Department of Physics and Astronomy, Iowa State University, Ames, Iowa, 50011, USA
| | - Apurba Lal Koner
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh, 462066, INDIA (TD) (ALK
| |
Collapse
|
37
|
Chen J, Li S, Ma D, Li L, Zhuang W, Chen M. A lipid droplet-specific fluorescence probe for atherosclerotic plaque imaging. Analyst 2022; 147:3081-3086. [PMID: 35678714 DOI: 10.1039/d1an01937f] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The dysregulation of lipid droplets (LDs) is closely related to certain metabolic diseases, while the role of LDs during pathological processes remains mysterious. It would be of great value to monitor the dynamic changes of LDs in a visible way so as to study their biological functions. In this study, we report a LD-specific fluorescence probe TBI for precise LD-targeting imaging in cells and atherosclerotic tissues. TBI exhibited great biocompatibility, remarkable oil-enhanced fluorescence emission, good photostability and impressive intracellular and tissular LD-specific imaging performance. Importantly, TBI could efficiently stain the LDs at a low concentration of 50 nM, and the motion tracking of LDs could be observed via fluorescence imaging. Moreover, TBI could efficiently light up the LD distribution in mouse atherosclerotic plaques with high resolution, which revealed the ultra-structure of atherosclerotic plaques. In conclusion, these results imply that TBI could be a potential tool for investigating the physiological and pathological role of LDs.
Collapse
Affiliation(s)
- Jingruo Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Shufen Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Di Ma
- Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Lilan Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Weihua Zhuang
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China.
| | - Mao Chen
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China. .,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, PR China.,Department of Cardiology, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu 610041, PR China
| |
Collapse
|
38
|
Jung Y, Jin JH, Kim Y, Oh JH, Moon H, Jeong H, Kim J, Park YK, Oh Y, Park S, Kim D. Development of a fluorescent nanoprobe based on an amphiphilic single-benzene-based fluorophore for lipid droplet detection and its practical applications. Org Biomol Chem 2022; 20:5423-5433. [PMID: 35758412 DOI: 10.1039/d2ob00830k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Lipid droplets (LDs) are crucial biological organelles connected with metabolic pathways in biological systems and diseases. To monitor the locations and accumulation of LDs in lipid-related diseases, the development of a visualization tool for LDs has gained importance. In particular, LD visualization using fluorescent probes has gained attention. Herein, a new fluorescent nanoprobe, BMeS-Ali, is developed that can sense LDs based on an amphiphilic single benzene-based fluorophore (SBBF). BMeS-Ali consists of hydrophilic (-NH2) and hydrophobic (-C12H25) moieties and exists as a micelle nanostructure in aqueous media. BMeS-Ali has a weak fluorescence, but its emission was dramatically enhanced upon exposure to the LD components such as oleic acids (OA) by reassembling its nano-formulation. BMeS-Ali showed a selective LD staining ability and great biocompatibility in cells (cancer cells and stem cells). It also showed a practical sensing ability towards biologically derived lipids and can be applied to the visualization of human fingerprints. We found that the nanoprobe BMeS-Ali has significant potential to serve as a practical dye and sensor for lipids, especially for LD imaging in the biomedical research area and broader industrial applications.
Collapse
Affiliation(s)
- Yuna Jung
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Ji Hye Jin
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Youngseo Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hyeon Oh
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Heechang Moon
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Huisu Jeong
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea
| | - Jaehoon Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Yoon Kyung Park
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Yohan Oh
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, Korea.,Department of Biochemistry and Molecular Biology, College of Medicine, Hanyang University, Seoul 04763, Korea
| | - Sungnam Park
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Dokyoung Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea. .,Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, Kyung Hee University, Seoul 02447, Republic of Korea.,Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.,Center for Converging Humanities, Kyung Hee University, Seoul 02447, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
39
|
Park ME, Lee KR, Chen GQ, Kim HU. Enhanced production of hydroxy fatty acids in Arabidopsis seed through modification of multiple gene expression. BIOTECHNOLOGY FOR BIOFUELS AND BIOPRODUCTS 2022; 15:66. [PMID: 35717237 PMCID: PMC9206371 DOI: 10.1186/s13068-022-02167-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 06/09/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND Castor (Ricinus communis L.) seeds contain unusual fatty acid, hydroxy fatty acid (HFA) used as a chemical feedstock for numerous industrial products. Castor cultivation is limited by the potent toxin ricin in its seeds and other poor agronomic traits, so it is advantageous to develop a suitable HFA-producing crop. Significant research efforts have been made to produce HFA in model Arabidopsis, but the level of HFA produced in transgenic Arabidopsis is much less than the level found in castor seeds which produce 90% HFA in seed oil. RESULTS We designed a transformation construct that allowed co-expression of five essential castor genes (named pCam5) involved in HFA biosynthesis, including an oleate [Formula: see text] 12-hydroxylase (FAH12), diacylglycerol (DAG) acyltransferase 2 (DGAT2), phospholipid: DAG acyltransferase 1-2 (PDAT1-2), phosphatidylcholine (PC): DAG cholinephosphotransferase (PDCT) and Lyso-PC acyltransferase (LPCAT). Transgenic Arabidopsis pCam5 lines produced HFA counting for 25% in seed oil. By knocking out Arabidopsis Fatty acid elongase 1 (AtFAE1) in pCam5 using CRISPR/Cas9 technology, the resulted pCam5-atfae1 lines produced over 31% of HFA. Astonishingly, the pCam5-atfae1 line increased seed size, weight, and total oil per seed exceeding wild type by 40%. Seed germination, seedling growth and seed mucilage content of pCam5-atfae1 lines were not affected by the genetic modification. CONCLUSIONS Our results provide not only insights for future research uncovering mechanisms of HFA synthesis in seed, but also metabolic engineering strategies for generating safe HFA-producing crops.
Collapse
Affiliation(s)
- Mid-Eum Park
- grid.263333.40000 0001 0727 6358Department of Molecular Biology, Sejong University, Seoul, Republic of Korea
| | - Kyeong-Ryeol Lee
- grid.420186.90000 0004 0636 2782Department of Agricultural Biotechnology, Rural Development Administration, National Institute of Agricultural Sciences, Jeonju, Republic of Korea
| | - Grace Q. Chen
- grid.417548.b0000 0004 0478 6311Western Regional Research Center, Agricultural Research Service, U.S. Department of Agriculture, Albany, CA USA
| | - Hyun Uk Kim
- grid.263333.40000 0001 0727 6358Department of Molecular Biology, Sejong University, Seoul, Republic of Korea ,grid.263333.40000 0001 0727 6358Department of Bioindustry and Bioresource Engineering, Plant Engineering Research Institute, Sejong University, Seoul, 05006 Republic of Korea
| |
Collapse
|
40
|
Kajiwara K, Osaki H, Greßies S, Kuwata K, Kim JH, Gensch T, Sato Y, Glorius F, Yamaguchi S, Taki M. A negative-solvatochromic fluorescent probe for visualizing intracellular distributions of fatty acid metabolites. Nat Commun 2022; 13:2533. [PMID: 35534485 PMCID: PMC9085894 DOI: 10.1038/s41467-022-30153-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 04/19/2022] [Indexed: 01/25/2023] Open
Abstract
Metabolic distribution of fatty acid to organelles is an essential biological process for energy homeostasis as well as for the maintenance of membrane integrity, and the metabolic pathways are strictly regulated in response to environmental stimuli. Herein, we report a fluorescent fatty acid probe, which bears an azapyrene dye that changes its absorption and emission features depending on the microenvironment polarity of the organelle into which it is transported. Owing to the environmental sensitivity of this dye, the distribution of the metabolically incorporated probe in non-polar lipid droplets, medium-polarity membranes, and the polar aqueous regions, can be visualized in different colors. Based on density scatter plots of the fluorophore, we demonstrate that the degradation of triacylglycerols in lipid droplets occurs predominantly via lipolysis rather than lipophagy in nutrition-starved hepatocytes. This tool can thus be expected to significantly advance our understanding of the lipid metabolism in living organisms. Metabolic distribution of fatty acids to organelles is an essential biological process for energy homeostasis. Here the authors report a fluorescent probe that allows multicolour visualisation of the intracellular distribution of exogenous fatty acids, metabolically incorporated as lipid components.
Collapse
|
41
|
Stone SJ. Mechanisms of intestinal triacylglycerol synthesis. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159151. [PMID: 35296424 DOI: 10.1016/j.bbalip.2022.159151] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/13/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023]
Abstract
Triacylglycerols are a major source of stored energy that are obtained either from the diet or can be synthesized to some extent by most tissues. Alterations in pathways of triacylglycerol metabolism can result in their excessive accumulation leading to obesity, insulin resistance, cardiovascular disease and nonalcoholic fatty liver disease. Most tissues in mammals synthesize triacylglycerols via the glycerol 3-phosphate pathway. However, in the small intestine the monoacylglycerol acyltransferase pathway is the predominant pathway for triacylglycerol biosynthesis where it participates in the absorption of dietary triacylglycerol. In this review, the enzymes that are part of both the glycerol 3-phosphate and monoacylglycerol acyltransferase pathways and their contributions to intestinal triacylglycerol metabolism are reviewed. The potential of some of the enzymes involved in triacylglycerol synthesis in the small intestine as possible therapeutic targets for treating metabolic disorders associated with elevated triacylglycerol is briefly discussed.
Collapse
Affiliation(s)
- Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
42
|
Paul B, Lewinska M, Andersen JB. Lipid alterations in chronic liver disease and liver cancer. JHEP Rep 2022; 4:100479. [PMID: 35469167 PMCID: PMC9034302 DOI: 10.1016/j.jhepr.2022.100479] [Citation(s) in RCA: 136] [Impact Index Per Article: 45.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 02/06/2023] Open
Abstract
Lipids are a complex and diverse group of molecules with crucial roles in many physiological processes, as well as in the onset, progression, and maintenance of cancers. Fatty acids and cholesterol are the building blocks of lipids, orchestrating these crucial metabolic processes. In the liver, lipid alterations are prevalent as a cause and consequence of chronic hepatitis B and C virus infections, alcoholic hepatitis, and non-alcoholic fatty liver disease and steatohepatitis. Recent developments in lipidomics have also revealed that dynamic changes in triacylglycerols, phospholipids, sphingolipids, ceramides, fatty acids, and cholesterol are involved in the development and progression of primary liver cancer. Accordingly, the transcriptional landscape of lipid metabolism suggests a carcinogenic role of increasing fatty acids and sterol synthesis. However, limited mechanistic insights into the complex nature of the hepatic lipidome have so far hindered the development of effective therapies.
Collapse
|
43
|
Wang Z, Li R, Chen L, Zhai X, Liu W, Lin X, Chen L, Chen N, Sun S, Li Z, Hao J, Chen X, Lin B, Xie L. Precise Molecular Design of a Pair of New Regioisomerized Fluorophores With Opposite Fluorescent Properties. Front Chem 2022; 9:823519. [PMID: 35127656 PMCID: PMC8811449 DOI: 10.3389/fchem.2021.823519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Aggregation-induced emission (AIE) has attracted much attention in the past 2 decades. To develop novel AIE-active materials, ACQ-to-AIE transformation via regioisomerization is one of the most straightforward method. However, most of the reported ACQ-to-AIE transformations are achieved by migrating bulky units. In this work, a facile conversion was realized by migrating a small pyrrolidinyl group from para- to ortho-position on the rofecoxib scaffold. As a result, a pair of new isomers named MOX2 and MOX4 exhibited AIE behavior and ACQ activity, respectively. Moreover, MOX2 also showed solvatochromic, mechanochromic, and acidochromic properties with reversible multi-stimulus behavior. Single crystal X-ray analysis of MOX2 revealed that the molecular conformation and its packing mode were responsible for the AIE emission behavior. Further investigation indicated that MOX2 showed high lipid droplets staining selectivity. Taken together, the current work not only provides a new design philosophy for achieving ACQ-to-AIE conversion by migrating a small pyrrolidinyl group but also presents a promising candidate MOX2 for potential applications such as in security ink, optical recording and biological applications.
Collapse
Affiliation(s)
- Zexin Wang
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, China
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Renfu Li
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Li Chen
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, China
| | - Xin Zhai
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Wei Liu
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, China
- School of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Xiang Lin
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, China
- Hengyang Medical School, Institute of Cytology and Genetics, University of South China, Hengyang, China
| | - Liwei Chen
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, China
| | - Nannan Chen
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, China
| | - Shitao Sun
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Zhenli Li
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Jinle Hao
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Xueyuan Chen
- CAS Key Laboratory of Design and Assembly of Functional Nanostructures, and Fujian Key Laboratory of Nanomaterials, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Bin Lin
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, China
| | - Lijun Xie
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, China
| |
Collapse
|
44
|
Li D, Xu N, Hou Y, Ren W, Zhang N, Wang X, Sun Y, Lu W, Qu G, Yu Y, Lv C, Han F. Abnormal lipid droplets accumulation induced cognitive deficits in obstructive sleep apnea syndrome mice via JNK/SREBP/ACC pathway but not through PDP1/PDC pathway. Mol Med 2022; 28:3. [PMID: 35030992 PMCID: PMC8760803 DOI: 10.1186/s10020-021-00427-8] [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: 09/06/2021] [Accepted: 12/17/2021] [Indexed: 11/25/2022] Open
Abstract
The mechanisms of chronic intermittent hypoxia (CIH)-induced cognitive deficits remain unclear. Here, our study found that about 3 months CIH treatment induced lipid droplets (LDs) accumulation in hippocampal nerve and glia cells of C57BL/6 mice, and caused severe neuro damage including neuron lesions, neuroblast (NB) apoptosis and abnormal glial activation. Studies have shown that the neuronal metabolism disorders might contribute to the CIH induced-hippocampal impairment. Mechanistically, the results showed that pyruvate dehydrogenase complex E1ɑ subunit (PDHA1) and the pyruvate dehydrogenase complex (PDC) activator pyruvate dehydrogenase phosphatase 1 (PDP1) did not noticeable change after intermittent hypoxia. Consistent with those results, the level of Acetyl-CoA in hippocampus did not significantly change after CIH exposure. Interestingly, we found that CIH produced large quantities of ROS, which activated the JNK/SREBP/ACC pathway in nerve and glia cells. ACC catalyzed the carboxylation of Acetyl-CoA to malonyl-CoA and then more lipid acids were synthesized, which finally caused aberrant LDs accumulation. Therefore, the JNK/SREBP/ACC pathway played a crucial role in the cognitive deficits caused by LDs accumulation after CIH exposure. Additionally, LDs were peroxidized by the high level of ROS under CIH conditions. Together, lipid metabolic disorders contributed to nerve and glia cells damage, which ultimately caused behavioral dysfunction. An active component of Salvia miltiorrhiza, SMND-309, dramatically alleviated these injuries and improved cognitive deficits of CIH mice.
Collapse
Affiliation(s)
- Dongze Li
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Na Xu
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Yanyan Hou
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Wenjing Ren
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Na Zhang
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Xi Wang
- Yantai Affiliated Hospital of Binzhou Medical University, 717 Jinbu Street, YanTai, 264199, China
| | - Yeying Sun
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Wenxue Lu
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Guiwu Qu
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China
| | - Yan Yu
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China.
| | - Changjun Lv
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China.
| | - Fang Han
- Binzhou Medical University, 346 Guanhai Road, YanTai, 264003, China.
| |
Collapse
|
45
|
Liu Y, Mihna D, Izem L, Morton RE. Both full length-cholesteryl ester transfer protein and exon 9-deleted cholesteryl ester transfer protein promote triacylglycerol storage in cultured hepatocytes. Lipids 2022; 57:69-79. [PMID: 34866179 PMCID: PMC9060302 DOI: 10.1002/lipd.12330] [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: 07/15/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
We previously reported that overexpression of full-length cholesteryl ester transfer protein (FL-CETP), but not its exon 9-deleted variant (∆E9-CETP), in an adipose cell line reduces their triacylglycerol (TAG) content. This provided mechanistic insight into several in vivo studies where FL-CETP levels are inversely correlated with adiposity. However, increased FL-CETP is also associated with elevated hepatic lipids, suggesting that the effect of CETP on cellular lipid metabolism may be tissue-specific. Here, we directly investigated the role of FL-CETP and ∆E9-CETP in hepatic lipid metabolism. FL- or ∆E9-CETP was overexpressed in HepG2-C3A by adenovirus transduction. Overexpression of either FL or ∆E9-CETP in hepatocytes increased cellular TAG mass by 25% but reduced TAG secretion. This cellular TAG was contained in larger and more numerous lipid droplets. Analysis of TAG synthetic and catabolic pathways showed that this elevated TAG content was due to increased incorporation of fatty acid into TAG (24%), and higher de novo synthesis of fatty acid (50%) and TAG from acetate (40%). siRNA knockdown of CETP had the opposite effect on TAG synthesis and lipogenesis, and decreased cellular TAG. This novel increase in cellular TAG by FL-CETP overexpression was reproduced in Caco-2 intestinal epithelial cells. We conclude that, unlike that seen in adipocyte cells, overexpression of either CETP isoform in lipoprotein-secreting cells promotes the accumulation of TAG. These data suggest that the in vivo correlation between CETP levels and hepatic steatosis can be explained, in part, by a direct effect of CETP on hepatocyte cellular metabolism.
Collapse
Affiliation(s)
- Yan Liu
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Daniel Mihna
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Lahoucine Izem
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Richard E Morton
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
46
|
McFie PJ, Chumala P, Katselis GS, Stone SJ. DGAT2 stability is increased in response to DGAT1 inhibition in gene edited HepG2 cells. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158991. [PMID: 34116261 DOI: 10.1016/j.bbalip.2021.158991] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 05/21/2021] [Accepted: 06/05/2021] [Indexed: 12/14/2022]
Abstract
In eukaryotic organisms, two unrelated acyl-CoA:diacylglycerol acyltransferase (DGAT) enzymes, DGAT1 and DGAT2, catalyze the final step of the triacylglycerol biosynthetic pathway. Both enzymes are highly expressed in lipogenic tissues, such as adipose tissue, small intestine and the liver. DGAT2 has a prominent role in hepatocyte lipid metabolism synthesizing triacylglycerols that are utilized for very low-density lipoprotein assembly. However, due to the lack of useful antibodies to detect endogenous DGAT2 protein, it has been difficult to determine how this enzyme functions at the cellular level. We have unsuccessfully tested many commercial antibodies as well as our own "in-house" antibodies. There is currently no evidence that DGAT2 undergoes processing such that antigenic epitopes to these antibodies are removed. As an alternative, many studies have utilized epitope tagged versions of DGAT2 overexpressed in cells. These approaches can provide valuable information about a protein, but can be subject to artifacts, such as mislocalization, misregulation, protein aggregation and abnormal protein-protein interactions. In this study, we used gene editing with CRISPR/Cas9 to add three consecutive FLAG epitopes to the C-terminus of endogenous DGAT2 in HepG2 cells. HepG2 cells, derived from a human hepatocellular carcinoma, have been routinely used as a cell model to study human hepatocyte lipid and lipoprotein metabolism. Using this system allowed us to successfully detect DGAT2 expressed from its endogenous locus in HepG2 cells by immunoblotting with anti-FLAG antibodies.
Collapse
Affiliation(s)
- Pamela J McFie
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada
| | - Paulos Chumala
- Department of Medicine and the Canadian Centre for Health and Safety in Agriculture, University of Saskatchewan, Saskatoon, Saskatchewan S7N 2Z4, Canada
| | - George S Katselis
- Department of Medicine and the Canadian Centre for Health and Safety in Agriculture, University of Saskatchewan, Saskatoon, Saskatchewan S7N 2Z4, Canada
| | - Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| |
Collapse
|
47
|
Cottier S, Schneiter R. Lipid droplets form a network interconnected by the endoplasmic reticulum through which their proteins equilibrate. J Cell Sci 2021; 135:271208. [PMID: 34373922 DOI: 10.1242/jcs.258819] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 07/03/2021] [Indexed: 01/13/2023] Open
Abstract
Lipid droplets (LDs) are globular intracellular structures dedicated to the storage of neutral lipids. They are closely associated with the endoplasmic reticulum (ER) and are delineated by a monolayer of phospholipids that is continuous with the cytoplasmic leaflet of the ER membrane. LDs contain a specific set of proteins, but how these proteins are targeted to the LD surface is not fully understood. Here, we devised a yeast mating-based microscopic readout to monitor the transfer of LD proteins upon zygote formation. The results of this analysis indicate that ER fusion between mating partners is required for transfer of LD proteins and that this transfer is continuous, bidirectional and affects most LDs simultaneously. These observations suggest that LDs do not fuse upon mating of yeast cells, but that they form a network that is interconnected through the ER membrane. Consistent with this, ER-localized LD proteins rapidly move onto LDs of a mating partner and this protein transfer is affected by seipin, a protein important for proper LD biogenesis and the functional connection of LDs with the ER membrane.
Collapse
Affiliation(s)
- Stéphanie Cottier
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| | - Roger Schneiter
- Department of Biology, University of Fribourg, Chemin du Musée 10, 1700 Fribourg, Switzerland
| |
Collapse
|
48
|
Reza AHMM, Zhou Y, Qin J, Tang Y. Aggregation-induced emission luminogens for lipid droplet imaging. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2021; 184:101-144. [PMID: 34749970 DOI: 10.1016/bs.pmbts.2021.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Lipid droplets (LDs) are evolutionarily conserved organelles involved in energy homeostasis and versatile intracellular processes in different cell types. Their importance is ubiquitous, ranges from utilization as the biofunctional components to third-generation biofuel production from microalgae, while morphology and functional perturbations could also relate to the multiple diseases in higher mammals. Biosynthesis of lipids can be triggered by multiple factors related to organismal physiology and the surrounding environment. An early prediction of this might help take necessary actions toward desired outcomes. In vivo visualization of LDs can give molecular insight into regulatory mechanisms and the underlying connections with other cellular structures. Traditional bioprobes for LDs detection often suffer from different dye-specific limitations such as aggregation-caused quenching and self-decomposition phenomena that hinder the research advancement. The emergence of lipid-specific nanoprobes with aggregation-induced emission (AIE) attributes in recent years is promising in remunerative characteristics with defined bioimaging properties. By utilizing the easy synthetic techniques and exploiting the unique physical features of these molecules, highly selective, stable, biocompatible and facile fluorescent probes could be fabricated for lipid detection. This chapter will provide up-to-date insight into the recent advances in lipid-specific AIE-based probes to enhance the opportunities for basic research related to the distinct roles of LDs in living organisms.
Collapse
Affiliation(s)
- A H M Mohsinul Reza
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia; Institute for NanoScale Science and Technology, College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| | - Yabin Zhou
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia
| | - Jianguang Qin
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia.
| | - Youhong Tang
- College of Science and Engineering, Flinders University, Adelaide, SA, Australia; Institute for NanoScale Science and Technology, College of Science and Engineering, Flinders University, Adelaide, SA, Australia.
| |
Collapse
|
49
|
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: 1.0] [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
|
50
|
Ralhan I, Chang CL, Lippincott-Schwartz J, Ioannou MS. Lipid droplets in the nervous system. J Cell Biol 2021; 220:e202102136. [PMID: 34152362 PMCID: PMC8222944 DOI: 10.1083/jcb.202102136] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 01/20/2023] Open
Abstract
Lipid droplets are dynamic intracellular lipid storage organelles that respond to the physiological state of cells. In addition to controlling cell metabolism, they play a protective role for many cellular stressors, including oxidative stress. Despite prior descriptions of lipid droplets appearing in the brain as early as a century ago, only recently has the role of lipid droplets in cells found in the brain begun to be understood. Lipid droplet functions have now been described for cells of the nervous system in the context of development, aging, and an increasing number of neuropathologies. Here, we review the basic mechanisms of lipid droplet formation, turnover, and function and discuss how these mechanisms enable lipid droplets to function in different cell types of the nervous system under healthy and pathological conditions.
Collapse
Affiliation(s)
- Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
| | - Chi-Lun Chang
- Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA
| | | | - Maria S. Ioannou
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, Alberta, Canada
- Department of Cell Biology, University of Alberta, Edmonton, Alberta, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|