1
|
Kim JJ, Yang EJ, Molina David J, Cho S, Ficarella M, Pape N, Schiffer JE, Njeim R, Kim SS, Lo Re C, Fontanella A, Kaber M, Sloan A, Merscher S, Fornoni A. Ezetimibe Enhances Lipid Droplet and Mitochondria Contact Formation, Improving Fatty Acid Transfer and Reducing Lipotoxicity in Alport Syndrome Podocytes. Int J Mol Sci 2024; 25:13134. [PMID: 39684843 DOI: 10.3390/ijms252313134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Mitochondrial dysfunction is a critical factor in the pathogenesis of Alport syndrome (AS), contributing to podocyte injury and disease progression. Ezetimibe, a lipid-lowering drug, is known to inhibit cholesterol and fatty acid uptake and to reduce triglyceride content in the kidney cortex of mice with AS. However, its effects on lipid droplet (LD) utilization by mitochondria have not been explored. Transmission electron microscopy (TEM) and mitochondrial functional assays (ATP production, mitochondrial membrane potential, and citrate synthase activity) were used to investigate the impact of ezetimibe on LD-mitochondria contact formation and mitochondrial function in Col4a3KO (AS) and wildtype (WT) podocytes. TEM analysis revealed significant mitochondrial abnormalities in AS podocytes, including swollen mitochondria and reduced cristae density, while mitochondrial function assays showed decreased ATP production and lowered mitochondrial membrane potential. AS podocytes also demonstrated a higher content of LD but with reduced LD-mitochondria contact sites. Ezetimibe treatment significantly increased the number of LD-mitochondria contact sites, enhanced fatty acid transfer efficiency, and reduced intracellular lipid accumulation. These changes were associated with a marked reduction in the markers of lipotoxicity, such as apoptosis and oxidative stress. Mitochondrial function was significantly improved, evidenced by increased basal respiration, ATP production, maximal respiration capacity, and the restoration of mitochondrial membrane potential. Additionally, mitochondrial swelling was significantly reduced in ezetimibe-treated AS podocytes. Our findings reveal a novel role for ezetimibe in enhancing LD-mitochondria contact formation, leading to more efficient fatty acid transfer, reduced lipotoxicity, and improved mitochondrial function in AS podocytes. These results suggest that ezetimibe could be a promising therapeutic agent for treating mitochondrial dysfunction and lipid metabolism abnormalities in AS.
Collapse
Affiliation(s)
- Jin-Ju Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Eun-Jeong Yang
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Judith Molina David
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sunjoo Cho
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Maria Ficarella
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Nils Pape
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Josephin Elizabeth Schiffer
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rachel Njeim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Stephanie S Kim
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Claudia Lo Re
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Unit of Nephrology and Dialysis, Department of Clinical and Experimental Medicine, A.O.U "G. Martino", University of Messina, 98122 Messina, Italy
| | - Antonio Fontanella
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Maria Kaber
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alexis Sloan
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sandra Merscher
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alessia Fornoni
- Katz Family Division of Nephrology and Hypertension, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Peggy and Harold Katz Family Drug Discovery Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
2
|
Wu Q, Jin C, Liu X, Zhang Q, Jiao B, Yu H. 1-Bromopropane induces mitochondrial damage and lipid metabolism imbalance in respiratory epithelial cells through the PGC-1α/PPARα pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 289:117492. [PMID: 39644563 DOI: 10.1016/j.ecoenv.2024.117492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/30/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
1-Bromopropane (1-BP) has become a new air pollutant in occupational and living environments due to its advantages in industrial applications and as a representative compound of volatile organic compounds (VOCs). As an irritant, its damaging effects on respiratory epithelium are worthy of further study. This study aimed to explore the damage effects of 1-BP on respiratory epithelial cells and reveal its underlying mechanisms. We found that exposure to 1-BP markedly reduced the viability of respiratory epithelial cells in a dose-dependent manner, and induced oxidative stress and vacuolation changes in respiratory epithelial cells. Subsequently, through RNA-seq analysis, we identified that the 1-BP-induced damage of respiratory epithelial cells was related to the mitochondrial function pathway and further verified that 1-BP caused mitochondrial damage of respiratory epithelial cells, which was manifested as ultrastructural damage, decreased membrane potential, ATP, and MFN2 levels. These damages were associated with cellular oxidative stress responses. Pretreating cells with the agonists of PGC-1α and PPARα, we revealed that 1-BP affected the expression of PGC-1α and interfered with its coactivator PPARα levels, causing an increase in the expression of lipid-producing genes and a decrease in the expression of lipid-decomposing genes, thus leading to a lipid accumulation in respiratory epithelial cells. Meanwhile, the imbalance of lipid metabolism in respiratory epithelial cells induced by 1-BP further caused mitochondrial damage, and the effect was bidirectional. These findings suggested that 1-BP has a potential role in inducing respiratory epithelial cell damage and is associated with the PGC-1α/PPARα signaling pathway.
Collapse
Affiliation(s)
- Qiuyun Wu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China; Key Laboratory of Human Genetics and Environmental Medicine, Xuzhou Medical University, Xuzhou 221004, China.
| | - Chunmeng Jin
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Xue Liu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Qianyi Zhang
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Biyang Jiao
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| | - Hongmin Yu
- School of Public Health, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
3
|
Zhao CZ, Ding HM, Hu ZQ, Zhou L, Du YQ, Zhou P, Wang L. Exploring the mechanism of Ling-Gui-Zhu-Gan decoction in metabolic cardiomyopathy via inhibiting ferroptosis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156234. [PMID: 39547098 DOI: 10.1016/j.phymed.2024.156234] [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: 06/26/2024] [Revised: 09/14/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVE This study was to investigate the mechanism of Ling-Gui-Zhu-Gan decoction (LGZGD) in regulating lipid metabolism and thus inhibiting ferroptosis. METHODS UPLC for the determination of the main chemical composition of LGZGD. A HF-induced rat model of metabolic cardiomyopathy was established. Echocardiography was used to detect cardiac function. Serum lipid levels, myocardial injury markers, and lipid peroxidation levels were detected. Pathological changes were detected. Lipid deposition was assessed by oil red O, and the mitochondrial ultrastructure was observed by electron microscopy. Mechanistically, PLIN5, CD36, ATGL, GPX4, ACSL4, FPN1, DRP1, MFF, FIS1, and OPA1 expressions were examined. After PA-induced H9c2 cells established, apoptosis, myocardial injury markers, and lipid peroxidation levels were detected and lipid deposition levels were assessed. The expressions of PLIN5, CD36, ATGL, GPX4, ACSL4 and FPN1 were detected. H9c2 cardiomyocytes with transient knockdown of PLIN5 and overexpression of PLIN5 were constructed and treated with drug administration and modeling, and the apoptosis level was detected by flow cytometry, the levels of lipid peroxidation and ROS were detected by fluorescence, and the protein and gene expressions of ACSL4 and GPX4 were detected. Results The main active components of LGZGD were liquiritin, isoliquiritin, cinnamic acid, cinnamaldehyde, glycyrrhizic acid, and atractylenolide III. LGZGD significantly improved cardiac dysfunction, lowered lipid level and lipid deposition, reduced CK, NT-proBNP and MDA levels, restored SOD levels, and improved inflammatory cell infiltration as well as collagen fiber deposition. LGZGD decreased the expression of PLIN5, CD36, ACSL4, and increased the expression of ATGL, GPX4, and FPN1. LGZGD also decreased the gene expression of DRP1, MFF, FIS1, and increased OPA1 expression. LGZGD significantly ameliorated PA-induced apoptosis, decreased lipid deposition, lowered lipid peroxidation levels and CK level, decreased PLIN5, CD36, and ACSL4 expressions, and increased ATGL, GPX4, and FPN1 expressions. LGZGD reversed cardiomyocyte injury aggravated by transient knockdown of PLIN5, decreased apoptosis levels, lipid peroxidation levels, ROS levels, and ACSL4 expressions, and increased GPX4 expression. LGZGD enhanced cardiomyocyte protection after overexpression of PLIN5, reduced apoptosis levels, lipid peroxidation level and ROS level, decreased ACSL4 expression, and increased GPX4 expression. CONCLUSION PLIN5 interferes with lipid peroxidation, regulates mitochondrial function, and inhibits HF-induced ferroptosis in cardiomyocytes. LGZGD ameliorates impairment of cardiac structural function in model rats through PLIN5-mediated ferroptosis pathway, and has the effect of preventing metabolic cardiomyopathy.
Collapse
Affiliation(s)
- Chuan-Zhi Zhao
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Hui-Min Ding
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Zi-Qing Hu
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Lan Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yong-Qin Du
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Peng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China; Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China; Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China.
| | - Liang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China; Research Institute of Integrated Traditional Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, Anhui, 230012, China; Anhui Provincial Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui, 230012, China.
| |
Collapse
|
4
|
Amari C, Carletti M, Yan S, Michaud M, Salvaing J. Lipid droplets degradation mechanisms from microalgae to mammals, a comparative overview. Biochimie 2024; 227:19-34. [PMID: 39299537 DOI: 10.1016/j.biochi.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/15/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Lipid droplets (LDs) are organelles composed of a hydrophobic core (mostly triacylglycerols and steryl esters) delineated by a lipid monolayer and found throughout the tree of life. LDs were seen for a long time as simple energy storage organelles but recent works highlighted their versatile roles in several fundamental cellular processes, particularly during stress response. LDs biogenesis occurs in the ER and their number and size can be dynamically regulated depending on their function, e.g. during development or stress. Understanding their biogenesis and degradation mechanisms is thus essential to better apprehend their roles. LDs degradation can occur in the cytosol by lipolysis or after their internalization into lytic compartments (e.g. vacuoles or lysosomes) using diverse mechanisms that depend on the considered organism, tissue, developmental stage or environmental condition. In this review, we summarize our current knowledge on the different LDs degradation pathways in several main phyla of model organisms, unicellular or pluricellular, photosynthetic or not (budding yeast, mammals, land plants and microalgae). We highlight the conservation of the main degradation pathways throughout evolution, but also the differences between organisms, or inside an organism between different organs. Finally, we discuss how this comparison can help to shed light on relationships between LDs degradation pathways and LDs functions.
Collapse
Affiliation(s)
- Chems Amari
- Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et Aux Energies Alternatives, IRIG, CEA-Grenoble, 17 Rue des Martyrs, 38000, Grenoble, France; Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris, France
| | - Marta Carletti
- Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et Aux Energies Alternatives, IRIG, CEA-Grenoble, 17 Rue des Martyrs, 38000, Grenoble, France
| | - Siqi Yan
- Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et Aux Energies Alternatives, IRIG, CEA-Grenoble, 17 Rue des Martyrs, 38000, Grenoble, France
| | - Morgane Michaud
- Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et Aux Energies Alternatives, IRIG, CEA-Grenoble, 17 Rue des Martyrs, 38000, Grenoble, France
| | - Juliette Salvaing
- Laboratoire de Physiologie Cellulaire et Végétale, Université Grenoble Alpes, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Centre National de la Recherche Scientifique, Commissariat à l'Energie Atomique et Aux Energies Alternatives, IRIG, CEA-Grenoble, 17 Rue des Martyrs, 38000, Grenoble, France.
| |
Collapse
|
5
|
Chen J, Markworth JF, Ferreira C, Zhang C, Kuang S. Lipid droplets as cell fate determinants in skeletal muscle. Trends Endocrinol Metab 2024:S1043-2760(24)00274-1. [PMID: 39613547 DOI: 10.1016/j.tem.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 12/01/2024]
Abstract
Lipid droplets (LDs) are dynamic organelles that communicate with other cellular components to orchestrate energetic homeostasis and signal transduction. In skeletal muscle, the presence and importance of LDs have been widely studied in myofibers of both rodents and humans under physiological conditions and in metabolic disorders. However, the role of LDs in myogenic stem cells has only recently begun to be unveiled. In this review we briefly summarize the process of LD biogenesis and degradation in the most prevalent model. We then review recent knowledge on LDs in skeletal muscle and muscle stem cells. We further introduce advanced methodologies for LD imaging and mass spectrometry that have propelled our understanding of the dynamics and heterogeneity of LDs.
Collapse
Affiliation(s)
- Jingjuan Chen
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA
| | - James F Markworth
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Christina Ferreira
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907, USA
| | - Chi Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Shihuan Kuang
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907, USA; Department of Orthopaedic Surgery, Duke University, Durham, NC 27710, USA; Purdue University Institute for Cancer Research, West Lafayette, IN 47907, USA.
| |
Collapse
|
6
|
Zanellati MC, Hsu CH, Cohen S. Imaging interorganelle contacts at a glance. J Cell Sci 2024; 137:jcs262020. [PMID: 39440475 PMCID: PMC11529887 DOI: 10.1242/jcs.262020] [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] [Indexed: 10/25/2024] Open
Abstract
Eukaryotic cells are compartmentalized into membrane-bound organelles that must coordinate their responses to stimuli. One way that organelles communicate is via membrane contact sites (MCSs), sites of close apposition between organelles used for the exchange of ions, lipids and information. In this Cell Science at a Glance article and the accompanying poster, we describe an explosion of new methods that have led to exciting progress in this area and discuss key examples of how these methods have advanced our understanding of MCSs. We discuss how diffraction-limited and super-resolution fluorescence imaging approaches have provided important insight into the biology of interorganelle communication. We also describe how the development of multiple proximity-based methods has enabled the detection of MCSs with high accuracy and precision. Finally, we assess how recent advances in electron microscopy (EM), considered the gold standard for detecting MCSs, have allowed the visualization of MCSs and associated proteins in 3D at ever greater resolution.
Collapse
Affiliation(s)
- Maria Clara Zanellati
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chih-Hsuan Hsu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
7
|
Corbo JH, Chung J. Mechanisms of lipid droplet degradation. Curr Opin Cell Biol 2024; 90:102402. [PMID: 39053179 DOI: 10.1016/j.ceb.2024.102402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
Lipid droplets (LDs) are subcellular organelles that play an integral role in lipid metabolism by regulating the storage and release of fatty acids, which are essential for energy production and various cellular processes. Lipolysis and lipophagy are the two major LD degradation pathways that mediate the utilization of lipids stored in these organelles. Recent studies have further uncovered alternative pathways, including direct lysosomal LD degradation and LD exocytosis. Here, we highlight recent findings that dissect the molecular basis of these diverse LD degradation pathways. Then, we discuss speculations on the crosstalk among these pathways and the potential unconventional roles of LD degradation.
Collapse
Affiliation(s)
- J H Corbo
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA
| | - J Chung
- Department of Molecular and Cellular Biology, Harvard University, 52 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
8
|
Ibayashi M, Tatsumi T, Tsukamoto S. Perilipin2 depletion causes lipid droplet enlargement in the ovarian corpus luteum in mice. J Reprod Dev 2024; 70:296-302. [PMID: 39010158 PMCID: PMC11461514 DOI: 10.1262/jrd.2024-023] [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: 03/05/2024] [Accepted: 06/17/2024] [Indexed: 07/17/2024] Open
Abstract
Lipid droplets (LDs) are endoplasmic reticulum-derived organelles that store neutral lipids (mostly triglycerides and cholesterol esters) within a phospholipid monolayer and appear in most eukaryotic cells. Perilipins (PLINs, comprising PLIN1-5) are abundant LD-associated proteins with highly variable expression levels among tissues. Although PLINs are expressed in the mammalian ovaries, little is known about their subcellular localization and physiological functions. In this study, we investigated the localization of PLIN1-3 and their relationship with LD synthesis using mCherry-HPos reporter mice, thereby enabling the visualization of LD biogenesis in vivo. PLIN2 and PLIN3 were localized as puncta in granulosa cells with low levels of LD synthesis in developing follicles. This localization pattern was quite different from that of PLIN1, which was mainly localized in the theca and interstitial cells with high levels of LD synthesis. In the corpus luteum, where LD synthesis is highly induced, PLIN2 and PLIN3 are abundant in the particulate structures, whereas PLIN1 is poorly distributed. We also generated global Plin2-deficient mice using the CRSPR/Cas9 system and demonstrated that the lack of PLIN2 did not alter the distribution of PLIN1 and PLIN3 but unexpectedly induced LD enlargement in the corpus luteum. Collectively, our results suggest that the localization of PLIN1-3 is spatiotemporally regulated and that PLIN2 deficiency influences LD mobilization in the corpus luteum within the ovaries.
Collapse
Affiliation(s)
- Megumi Ibayashi
- Laboratory Animal and Bioresource Sciences Section, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| | - Takayuki Tatsumi
- Division of Reproductive Medicine, Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo 157-8535, Japan
| | - Satoshi Tsukamoto
- Laboratory Animal and Bioresource Sciences Section, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan
| |
Collapse
|
9
|
Klemm RW, Carvalho P. Lipid Droplets Big and Small: Basic Mechanisms That Make Them All. Annu Rev Cell Dev Biol 2024; 40:143-168. [PMID: 39356808 DOI: 10.1146/annurev-cellbio-012624-031419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
Lipid droplets (LDs) are dynamic storage organelles with central roles in lipid and energy metabolism. They consist of a core of neutral lipids, such as triacylglycerol, which is surrounded by a monolayer of phospholipids and specialized surface proteins. The surface composition determines many of the LD properties, such as size, subcellular distribution, and interaction with partner organelles. Considering the diverse energetic and metabolic demands of various cell types, it is not surprising that LDs are highly heterogeneous within and between cell types. Despite their diversity, all LDs share a common biogenesis mechanism. However, adipocytes have evolved specific adaptations of these basic mechanisms, enabling the regulation of lipid and energy metabolism at both the cellular and organismal levels. Here, we discuss recent advances in the understanding of both the general mechanisms of LD biogenesis and the adipocyte-specific adaptations controlling these fascinating organelles.
Collapse
Affiliation(s)
- Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom;
| | - Pedro Carvalho
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom;
| |
Collapse
|
10
|
Gamuyao R, Chang CL. Imaging and proteomics toolkits for studying organelle contact sites. Front Cell Dev Biol 2024; 12:1466915. [PMID: 39381373 PMCID: PMC11458464 DOI: 10.3389/fcell.2024.1466915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/05/2024] [Indexed: 10/10/2024] Open
Abstract
Organelle contact sites are regions where two heterologous membranes are juxtaposed by molecular tethering complexes. These contact sites are important in inter-organelle communication and cellular functional integration. However, visualizing these minute foci and identifying contact site proteomes have been challenging. In recent years, fluorescence-based methods have been developed to visualize the dynamic physical interaction of organelles while proximity labeling approaches facilitate the profiling of proteomes at contact sites. In this review, we explain the design principle for these contact site reporters: a dual-organelle interaction mechanism based on how endogenous tethers and/or tethering complexes localize to contact sites. We classify the contact site reporters into three categories: (i) single-protein systems, (ii) two-component systems with activated reporter signal upon organelle proximity, and (iii) reporters for contact site proteomes. We also highlight advanced imaging analysis with high temporal-spatial resolution and the use of machine-learning algorithms for detecting contact sites.
Collapse
Affiliation(s)
| | - Chi-Lun Chang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|
11
|
Miner GE, Cohen S. Protocol for monitoring fatty acid trafficking from lipid droplets to mitochondria in cultured cells. STAR Protoc 2024; 5:103236. [PMID: 39146189 PMCID: PMC11372805 DOI: 10.1016/j.xpro.2024.103236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/11/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024] Open
Abstract
Intracellular trafficking of fatty acids (FAs) between organelles is critical for cells to adjust their metabolism in response to stimuli such as exercise, fasting, and cold exposure. Here, we describe a protocol to monitor trafficking of FAs from lipid droplets to mitochondria. We describe the labeling of organelles in cultured C2C12 myoblasts with transfection and dyes. We detail a pulse-chase labeling paradigm using a fluorescent FA analog, live-cell imaging to visualize trafficking of FAs, and steps to quantify FA trafficking. For complete details on the use and execution of this protocol, please refer to Miner et al.1.
Collapse
Affiliation(s)
- Gregory E Miner
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
12
|
Li X, Gamuyao R, Wu ML, Cho WJ, King SV, Petersen R, Stabley DR, Lindow C, Climer LK, Shirinifard A, Ferrara F, Throm RE, Robinson CG, Zhou Y, Carisey AF, Tebo AG, Chang CL. A fluorogenic complementation tool kit for interrogating lipid droplet-organelle interaction. J Cell Biol 2024; 223:e202311126. [PMID: 38949658 PMCID: PMC11215687 DOI: 10.1083/jcb.202311126] [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] [Received: 11/22/2023] [Revised: 04/24/2024] [Accepted: 05/31/2024] [Indexed: 07/02/2024] Open
Abstract
Contact sites between lipid droplets and other organelles are essential for cellular lipid and energy homeostasis upon metabolic demands. Detection of these contact sites at the nanometer scale over time in living cells is challenging. We developed a tool kit for detecting contact sites based on fluorogen-activated bimolecular complementation at CONtact sites, FABCON, using a reversible, low-affinity split fluorescent protein, splitFAST. FABCON labels contact sites with minimal perturbation to organelle interaction. Via FABCON, we quantitatively demonstrated that endoplasmic reticulum (ER)- and mitochondria (mito)-lipid droplet contact sites are dynamic foci in distinct metabolic conditions, such as during lipid droplet biogenesis and consumption. An automated analysis pipeline further classified individual contact sites into distinct subgroups based on size, likely reflecting differential regulation and function. Moreover, FABCON is generalizable to visualize a repertoire of organelle contact sites including ER-mito. Altogether, FABCON reveals insights into the dynamic regulation of lipid droplet-organelle contact sites and generates new hypotheses for further mechanistical interrogation during metabolic regulation.
Collapse
Affiliation(s)
- Xiao Li
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Rico Gamuyao
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Ming-Lun Wu
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Woo Jung Cho
- Cell and Tissue Imaging Center, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sharon V. King
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - R.A. Petersen
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Daniel R. Stabley
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Caleb Lindow
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Leslie K. Climer
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Francesca Ferrara
- Vector Production and Development Laboratory, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Robert E. Throm
- Vector Production and Development Laboratory, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Camenzind G. Robinson
- Cell and Tissue Imaging Center, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Yiwang Zhou
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Alexandre F. Carisey
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Alison G. Tebo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Chi-Lun Chang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| |
Collapse
|
13
|
Cardaci TD, VanderVeen BN, Huss AR, Bullard BM, Velázquez KT, Frizzell N, Carson JA, Price RL, Murphy EA. Decreased skeletal muscle intramyocellular lipid droplet-mitochondrial contact contributes to myosteatosis in cancer cachexia. Am J Physiol Cell Physiol 2024; 327:C684-C697. [PMID: 39010842 PMCID: PMC11427022 DOI: 10.1152/ajpcell.00345.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/20/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
Cancer cachexia, the unintentional loss of lean mass, contributes to functional dependency, poor treatment outcomes, and decreased survival. Although its pathogenicity is multifactorial, metabolic dysfunction remains a hallmark of cachexia. However, significant knowledge gaps exist in understanding the role of skeletal muscle lipid metabolism and dynamics in this condition. We examined skeletal muscle metabolic dysfunction, intramyocellular lipid droplet (LD) content, LD morphology and subcellular distribution, and LD-mitochondrial interactions using the Lewis lung carcinoma (LLC) murine model of cachexia. C57/BL6 male mice (n = 20) were implanted with LLC cells (106) in the right flank or underwent PBS sham injections. Skeletal muscle was excised for transmission electron microscopy (TEM; soleus), oil red O/lipid staining [tibialis anterior (TA)], and protein (gastrocnemius). LLC mice had a greater number (232%; P = 0.006) and size (130%; P = 0.023) of intramyocellular LDs further supported by increased oil-red O positive (87%; P = 0.0109) and "very high" oil-red O positive (178%; P = 0.0002) fibers compared with controls and this was inversely correlated with fiber size (R2 = 0.5294; P < 0.0001). Morphological analyses of LDs show increased elongation and complexity [aspect ratio: intermyofibrillar (IMF) = 9%, P = 0.046) with decreases in circularity [circularity: subsarcolemmal (SS) = 6%, P = 0.042] or roundness (roundness: whole = 10%, P = 0.033; IMF = 8%, P = 0.038) as well as decreased LD-mitochondria touch (-15%; P = 0.006), contact length (-38%; P = 0.036), and relative contact (86%; P = 0.004). Furthermore, dysregulation in lipid metabolism (adiponectin, CPT1b) and LD-associated proteins, perilipin-2 and perilipin-5, in cachectic muscle (P < 0.05) were observed. Collectively, we provide evidence that skeletal muscle myosteatosis, altered LD morphology, and decreased LD-mitochondrial interactions occur in a preclinical model of cancer cachexia.NEW & NOTEWORTHY We sought to advance our understanding of skeletal muscle lipid metabolism and dynamics in cancer cachexia. Cachexia increased the number and size of intramyocellular lipid droplets (LDs). Furthermore, decreases in LD-mitochondrial touch, contact length, and relative contact along with increased LD shape complexity with decreases in circularity and roundness. Dysregulation in lipid metabolism and LD-associated proteins was also documented. Collectively, we show that myosteatosis, altered LD morphology, and decreased LD-mitochondrial interactions occur in cancer cachexia.
Collapse
Affiliation(s)
- Thomas D Cardaci
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Brandon N VanderVeen
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Alexander R Huss
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Brooke M Bullard
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States
- Columbia Department of Veterans Affairs Health Care System, Columbia, South Carolina, United States
| | - Norma Frizzell
- Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - James A Carson
- Department of Kinesiology and Sports Management, JL Huffines Institute for Sports Medicine & Human Performance, Texas A&M University, College Station, Texas, United States
| | - Robert L Price
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - E Angela Murphy
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| |
Collapse
|
14
|
Boone C, Lewis SC. Bridging lipid metabolism and mitochondrial genome maintenance. J Biol Chem 2024; 300:107498. [PMID: 38944117 PMCID: PMC11326895 DOI: 10.1016/j.jbc.2024.107498] [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: 12/19/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/01/2024] Open
Abstract
Mitochondria are the nexus of cellular energy metabolism and major signaling hubs that integrate information from within and without the cell to implement cell function. Mitochondria harbor a distinct polyploid genome, mitochondrial DNA (mtDNA), that encodes respiratory chain components required for energy production. MtDNA mutation and depletion have been linked to obesity and metabolic syndrome in humans. At the cellular and subcellular levels, mtDNA synthesis is coordinated by membrane contact sites implicated in lipid transfer from the endoplasmic reticulum, tying genome maintenance to lipid storage and homeostasis. Here, we examine the relationship between mtDNA and lipid trafficking, the influence of lipotoxicity on mtDNA integrity, and how lipid metabolism may be disrupted in primary mtDNA disease.
Collapse
Affiliation(s)
- Casadora Boone
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, USA
| | - Samantha C Lewis
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, California, USA; Department of Molecular and Cell Biology, University of California, Berkeley, California, USA.
| |
Collapse
|
15
|
Emerson JI, Shi W, Conlon FL. Sex-Specific Response to A1BG Loss Results in Female Dilated Cardiomyopathy. RESEARCH SQUARE 2024:rs.3.rs-4631369. [PMID: 39070637 PMCID: PMC11276010 DOI: 10.21203/rs.3.rs-4631369/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Cardiac disease often manifests differently in terms of frequency and pathology between men and women. However, the mechanisms underlying these differences are not fully understood. The glycoprotein A1BG is necessary for proper cardiac function in females but not males. Despite this, the role of A1BG in the female heart remains poorly studied. Methods To determine the sex differential function of A1BG, we generated a novel conditional A1bg allele and a novel conditional A1bg Rosa26 knockin allele. Histology, electrocardiography, transcriptional profiling (RNA-seq), transmission electron microscopy, western blot analyses, mass spectrometry, and immunohistochemistry were used to assess cardiac structure and function. Results The study reveals that the absence of A1BG results in significant cardiac dysfunction in female but not male mice. Gene expression underscores that A1BG plays a critical role in metabolic processes and the integrity of intercalated discs in female cardiomyocytes. This dysfunction may be related to sex-specific A1BG cardiac interactomes and manifests as structural and functional alterations in the left ventricle indicative of dilated cardiomyopathy, thus suggesting a sex-specific requirement for A1BG in cardiac health. Conclusion The loss of A1BG in cardiomyocytes leads to dilated cardiomyopathy in females, not males.
Collapse
Affiliation(s)
| | - Wei Shi
- University of North Carolina at Chapel Hill
| | | |
Collapse
|
16
|
Fan H, Tan Y. Lipid Droplet-Mitochondria Contacts in Health and Disease. Int J Mol Sci 2024; 25:6878. [PMID: 38999988 PMCID: PMC11240910 DOI: 10.3390/ijms25136878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/19/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
The orchestration of cellular metabolism and redox balance is a complex, multifaceted process crucial for maintaining cellular homeostasis. Lipid droplets (LDs), once considered inert storage depots for neutral lipids, are now recognized as dynamic organelles critical in lipid metabolism and energy regulation. Mitochondria, the powerhouses of the cell, play a central role in energy production, metabolic pathways, and redox signaling. The physical and functional contacts between LDs and mitochondria facilitate a direct transfer of lipids, primarily fatty acids, which are crucial for mitochondrial β-oxidation, thus influencing energy homeostasis and cellular health. This review highlights recent advances in understanding the mechanisms governing LD-mitochondria interactions and their regulation, drawing attention to proteins and pathways that mediate these contacts. We discuss the physiological relevance of these interactions, emphasizing their role in maintaining energy and redox balance within cells, and how these processes are critical in response to metabolic demands and stress conditions. Furthermore, we explore the pathological implications of dysregulated LD-mitochondria interactions, particularly in the context of metabolic diseases such as obesity, diabetes, and non-alcoholic fatty liver disease, and their potential links to cardiovascular and neurodegenerative diseases. Conclusively, this review provides a comprehensive overview of the current understanding of LD-mitochondria interactions, underscoring their significance in cellular metabolism and suggesting future research directions that could unveil novel therapeutic targets for metabolic and degenerative diseases.
Collapse
Affiliation(s)
- Hongjun Fan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| | - Yanjie Tan
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, Collaborative Innovation Center of Cell Biology in Universities of Shandong, College of Life Sciences, Shandong Normal University, Jinan 250014, China
| |
Collapse
|
17
|
Wang Y, Zeng D, Wei L, Chen J, Li H, Wen L, Huang G, Dai Z, Luo J, Sun J, Xi Q, Zhang Y, Chen T. Effects of emulsifiers on lipid metabolism and performance of yellow-feathered broilers. BMC Vet Res 2024; 20:246. [PMID: 38849831 PMCID: PMC11157903 DOI: 10.1186/s12917-024-04095-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Reducing production costs while producing high-quality livestock and poultry products is an ongoing concern in the livestock industry. The addition of oil to livestock and poultry diets can enhance feed palatability and improve growth performance. Emulsifiers can be used as potential feed supplements to improve dietary energy utilization and maintain the efficient productivity of broilers. Therefore, further investigation is warranted to evaluate whether dietary emulsifier supplementation can improve the efficiency of fat utilization in the diet of yellow-feathered broilers. In the present study, the effects of adding emulsifier to the diet on lipid metabolism and the performance of yellow-feathered broilers were tested. A total of 240 yellow-feasted broilers (21-day-old) were randomly divided into 4 groups (6 replicates per group, 10 broilers per replicate, half male and half female within each replicate). The groups were as follows: the control group (fed with basal diet), the group fed with basal diet supplemented with 500 mg/kg emulsifier, the group fed with a reduced oil diet (reduced by 1%) supplemented with 500 mg/kg emulsifier, and the group fed with a reduced oil diet supplemented with 500 mg/kg emulsifier. The trial lasted for 42 days, during which the average daily feed intake, average daily gain, and feed-to-gain ratio were measured. Additionally, the expression levels of lipid metabolism-related genes in the liver, abdominal fat and each intestinal segment were assessed. RESULTS The results showed that compared with the basal diet group, (1) The average daily gain of the basal diet + 500 mg/kg emulsifier group significantly increased (P < 0.05), and the half-even-chamber rate was significantly increased (P < 0.05); (2) The mRNA expression levels of Cd36, Dgat2, Apob, Fatp4, Fabp2, and Mttp in the small intestine were significantly increased (P < 0.05). (3) Furthermore, liver TG content significantly decreased (P < 0.05), and the mRNA expression level of Fasn in liver was significantly decreased (P < 0.05), while the expression of Apob, Lpl, Cpt-1, and Pparα significantly increased (P < 0.05). (4) The mRNA expression levels of Lpl and Fatp4 in adipose tissue were significantly increased (P < 0.05), while the expression of Atgl was significantly decreased (P < 0.05). (5) Compared with the reduced oil diet group, the half-evading rate and abdominal fat rate of broilers in the reduced oil diet + 500 mg/kg emulsifier group were significantly increased (P < 0.05), and the serum level of LDL-C increased significantly (P < 0.05)0.6) The mRNA expression levels of Cd36, Fatp4, Dgat2, Apob, and Mttp in the small intestine were significantly increased (P < 0.05). 7) The mRNA expression levels of Fasn and Acc were significantly decreased in the liver (P < 0.05), while the mRNA expression levels of Lpin1, Dgat2, Apob, Lpl, Cpt-1, and Pparα were significantly increased (P < 0.05). CONCLUSIONS These results suggest that dietary emulsifier can enhance the fat utilization efficiency of broilers by increasing the small intestinal fatty acid uptake capacity, inhibiting hepatic fatty acid synthesis and promoting hepatic TG synthesis and transport capacity. This study provides valuable insights for the potential use of emulsifier supplementation to improve the performance of broiler chickens.
Collapse
Affiliation(s)
- Yuxuan Wang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Dewei Zeng
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Limin Wei
- Hainan Key Laboratory of Tropical Animal Breeding and Epidemic Research, Institute of Animal Husbandry and Veterinary Research, Hainan Academy of Agricultural Sciences, Haikou, Hainan, 571100, China
| | - Jingshen Chen
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Hongyi Li
- Yingdong College of Biology and Agriculture, Shaoguan University, Shaoguan, Guangdong, 512005, China
| | - Lijun Wen
- Guangdong Hainachuan Biotechnology Co., LTD, Guangzhou, Guangdong, 528515, China
| | - Guangming Huang
- Guangdong Hainachuan Biotechnology Co., LTD, Guangzhou, Guangdong, 528515, China
| | - Zhenqing Dai
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Junyi Luo
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jiajie Sun
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qianyun Xi
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Yongliang Zhang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Ting Chen
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, National Engineering Research Center for Breeding Swine Industry, State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
18
|
Meng Y, Guo D, Lin L, Zhao H, Xu W, Luo S, Jiang X, Li S, He X, Zhu R, Shi R, Xiao L, Wu Q, He H, Tao J, Jiang H, Wang Z, Yao P, Xu D, Lu Z. Glycolytic enzyme PFKL governs lipolysis by promoting lipid droplet-mitochondria tethering to enhance β-oxidation and tumor cell proliferation. Nat Metab 2024; 6:1092-1107. [PMID: 38773347 DOI: 10.1038/s42255-024-01047-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/10/2024] [Indexed: 05/23/2024]
Abstract
Lipid droplet tethering with mitochondria for fatty acid oxidation is critical for tumor cells to counteract energy stress. However, the underlying mechanism remains unclear. Here, we demonstrate that glucose deprivation induces phosphorylation of the glycolytic enzyme phosphofructokinase, liver type (PFKL), reducing its activity and favoring its interaction with perilipin 2 (PLIN2). On lipid droplets, PFKL acts as a protein kinase and phosphorylates PLIN2 to promote the binding of PLIN2 to carnitine palmitoyltransferase 1A (CPT1A). This results in the tethering of lipid droplets and mitochondria and the recruitment of adipose triglyceride lipase to the lipid droplet-mitochondria tethering regions to engage lipid mobilization. Interfering with this cascade inhibits tumor cell proliferation, promotes apoptosis and blunts liver tumor growth in male mice. These results reveal that energy stress confers a moonlight function to PFKL as a protein kinase to tether lipid droplets with mitochondria and highlight the crucial role of PFKL in the integrated regulation of glycolysis, lipid metabolism and mitochondrial oxidation.
Collapse
Affiliation(s)
- Ying Meng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Dong Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liming Lin
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hong Zhao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weiting Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shudi Luo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoming Jiang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shan Li
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xuxiao He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Rongxuan Zhu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Rongkai Shi
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Liwei Xiao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qingang Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haiyan He
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jingjing Tao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongfei Jiang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Zheng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| | - Pengbo Yao
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Daqian Xu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
- Institute of Fundamental and Transdisciplinary Research, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
19
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and Tgfβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. eLife 2024; 13:e95640. [PMID: 38805545 PMCID: PMC11132684 DOI: 10.7554/elife.95640] [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: 01/02/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that CBFB (subunit of a heterodimeric Cbfβ/Runx1, Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfb in tamoxifen-induced Cbfbf/f;Col2a1-CreERT mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfβ deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/Yap signaling and Tgfβ signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfb overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfb overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfβ may be the cause of OA. Moreover, Local admission of Cbfb may rescue and protect OA through decreasing Wnt/β-catenin signaling, and increasing Hippo/Yap signaling and Tgfβ/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfb overexpression could be an effective strategy for treatment of OA.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane UniversityNew OrleansUnited States
- Department of Pathology, School of Medicine, University of Alabama at BirminghamBirminghamUnited States
| |
Collapse
|
20
|
Zhao J, Li L, Wang X, Shen J. KN-93 promotes HDAC4 nucleus translocation to promote fatty acid oxidation in myocardial infarction. Exp Cell Res 2024; 438:114050. [PMID: 38663474 DOI: 10.1016/j.yexcr.2024.114050] [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: 09/30/2023] [Revised: 02/23/2024] [Accepted: 04/13/2024] [Indexed: 04/30/2024]
Abstract
Myocardial infarction (MI) is a potentially fatal disease that causes a significant number of deaths worldwide. The strategy of increasing fatty acid oxidation in myocytes is considered a therapeutic avenue to accelerate metabolism to meet energy demands. We conducted the study aiming to investigate the effect of KN-93, which induces histone deacetylase (HDAC)4 shuttling to the nucleus, on fatty acid oxidation and the expression of related genes. A mouse model of myocardial infarction was induced by isoprenaline administration. Heart damage was assessed by the detection of cardiac injury markers. The level of fatty acid oxidation level was evaluated by testing the expression of related genes. Both immunofluorescence and immunoblotting in the cytosol or nucleus were utilized to observe the distribution of HDAC4. The interaction between HDAC4 and specificity protein (SP)1 was confirmed by co-immunoprecipitation. The acetylation level of SP1 was tested after KN-93 treatment and HDAC4 inhibitor. Oxygen consumption rate and immunoblotting experiments were used to determine whether the effect of KN-93 on increasing fatty acid oxidation is through HDAC4 and SP1. Administration of KN-93 significantly reduced cardiac injury in myocardial infarction and promoted fatty acid oxidation both in vitro and in vivo. KN-93 was shown to mediate nuclear translocation of HDAC4. HDAC4 was found to interact with SP1 and reduce SP1 acetylation. HDAC4 or SP1 inhibitors attenuated the effect of KN-93 on fatty acid oxidation. In conclusion, KN-93 promotes HDAC4 translocation to the nucleus, thereby potentially enhancing fatty acid oxidation by SP1.
Collapse
Affiliation(s)
- Jianqiao Zhao
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, China
| | - Luona Li
- Department of Gastronomy, Nanjing Drum Tower Hospital, China
| | - Xindong Wang
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, China
| | - Jianping Shen
- Department of Cardiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, China.
| |
Collapse
|
21
|
Saukko-Paavola AJ, Klemm RW. Remodelling of mitochondrial function by import of specific lipids at multiple membrane-contact sites. FEBS Lett 2024; 598:1274-1291. [PMID: 38311340 DOI: 10.1002/1873-3468.14813] [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: 11/29/2023] [Revised: 12/14/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024]
Abstract
Organelles form physical and functional contact between each other to exchange information, metabolic intermediates, and signaling molecules. Tethering factors and contact site complexes bring partnering organelles into close spatial proximity to establish membrane contact sites (MCSs), which specialize in unique functions like lipid transport or Ca2+ signaling. Here, we discuss how MCSs form dynamic platforms that are important for lipid metabolism. We provide a perspective on how import of specific lipids from the ER and other organelles may contribute to remodeling of mitochondria during nutrient starvation. We speculate that mitochondrial adaptation is achieved by connecting several compartments into a highly dynamic organelle network. The lipid droplet appears to be a central hub in coordinating the function of these organelle neighborhoods.
Collapse
Affiliation(s)
| | - Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK
| |
Collapse
|
22
|
Enkler L, Spang A. Functional interplay of lipid droplets and mitochondria. FEBS Lett 2024; 598:1235-1251. [PMID: 38268392 DOI: 10.1002/1873-3468.14809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/12/2023] [Accepted: 01/04/2024] [Indexed: 01/26/2024]
Abstract
Our body stores energy mostly in form of fatty acids (FAs) in lipid droplets (LDs). From there the FAs can be mobilized and transferred to peroxisomes and mitochondria. This transfer is dependent on close opposition of LDs and mitochondria and peroxisomes and happens at membrane contact sites. However, the composition and the dynamics of these contact sites is not well understood, which is in part due to the dependence on the metabolic state of the cell and on the cell- and tissue-type. Here, we summarize the current knowledge on the contacts between lipid droplets and mitochondria both in mammals and in the yeast Saccharomyces cerevisiae, in which various contact sites are well studied. We discuss possible functions of the contact site and their implication in disease.
Collapse
Affiliation(s)
| | - Anne Spang
- Biozentrum, University of Basel, Switzerland
| |
Collapse
|
23
|
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
|
24
|
Griseti E, Bello AA, Bieth E, Sabbagh B, Iacovoni JS, Bigay J, Laurell H, Čopič A. Molecular mechanisms of perilipin protein function in lipid droplet metabolism. FEBS Lett 2024; 598:1170-1198. [PMID: 38140813 DOI: 10.1002/1873-3468.14792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 11/27/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023]
Abstract
Perilipins are abundant lipid droplet (LD) proteins present in all metazoans and also in Amoebozoa and fungi. Humans express five perilipins, which share a similar domain organization: an amino-terminal PAT domain and an 11-mer repeat region, which can fold into amphipathic helices that interact with LDs, followed by a structured carboxy-terminal domain. Variations of this organization that arose during vertebrate evolution allow for functional specialization between perilipins in relation to the metabolic needs of different tissues. We discuss how different features of perilipins influence their interaction with LDs and their cellular targeting. PLIN1 and PLIN5 play a direct role in lipolysis by regulating the recruitment of lipases to LDs and LD interaction with mitochondria. Other perilipins, particularly PLIN2, appear to protect LDs from lipolysis, but the molecular mechanism is not clear. PLIN4 stands out with its long repetitive region, whereas PLIN3 is most widely expressed and is used as a nascent LD marker. Finally, we discuss the genetic variability in perilipins in connection with metabolic disease, prominent for PLIN1 and PLIN4, underlying the importance of understanding the molecular function of perilipins.
Collapse
Affiliation(s)
- Elena Griseti
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Abdoul Akim Bello
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Eric Bieth
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
- Departement de Génétique Médicale, Centre Hospitalier Universitaire de Toulouse, France
| | - Bayane Sabbagh
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| | - Jason S Iacovoni
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Joëlle Bigay
- Institut de Pharmacologie Moléculaire et Cellulaire - IPMC, Université Côte d'Azur, CNRS, Valbonne, France
| | - Henrik Laurell
- Institut des Maladies Métaboliques et Cardiovasculaires - I2MC, Université de Toulouse, Inserm, Université Toulouse III - Paul Sabatier (UPS), France
| | - Alenka Čopič
- Centre de Recherche en Biologie Cellulaire de Montpellier - CRBM, Université de Montpellier, CNRS, France
| |
Collapse
|
25
|
Nakamura M. Lipotoxicity as a therapeutic target in obesity and diabetic cardiomyopathy. JOURNAL OF PHARMACY & PHARMACEUTICAL SCIENCES : A PUBLICATION OF THE CANADIAN SOCIETY FOR PHARMACEUTICAL SCIENCES, SOCIETE CANADIENNE DES SCIENCES PHARMACEUTIQUES 2024; 27:12568. [PMID: 38706718 PMCID: PMC11066298 DOI: 10.3389/jpps.2024.12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/09/2024] [Indexed: 05/07/2024]
Abstract
Unhealthy sources of fats, ultra-processed foods with added sugars, and a sedentary lifestyle make humans more susceptible to developing overweight and obesity. While lipids constitute an integral component of the organism, excessive and abnormal lipid accumulation that exceeds the storage capacity of lipid droplets disrupts the intracellular composition of fatty acids and results in the release of deleterious lipid species, thereby giving rise to a pathological state termed lipotoxicity. This condition induces endoplasmic reticulum stress, mitochondrial dysfunction, inflammatory responses, and cell death. Recent advances in omics technologies and analytical methodologies and clinical research have provided novel insights into the mechanisms of lipotoxicity, including gut dysbiosis, epigenetic and epitranscriptomic modifications, dysfunction of lipid droplets, post-translational modifications, and altered membrane lipid composition. In this review, we discuss the recent knowledge on the mechanisms underlying the development of lipotoxicity and lipotoxic cardiometabolic disease in obesity, with a particular focus on lipotoxic and diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Michinari Nakamura
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, United States
| |
Collapse
|
26
|
Kim H, Oh S, Lee S, Lee KS, Park Y. Recent advances in label-free imaging and quantification techniques for the study of lipid droplets in cells. Curr Opin Cell Biol 2024; 87:102342. [PMID: 38428224 DOI: 10.1016/j.ceb.2024.102342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/20/2024] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
Lipid droplets (LDs), once considered mere storage depots for lipids, have gained recognition for their intricate roles in cellular processes, including metabolism, membrane trafficking, and disease states like obesity and cancer. This review explores label-free imaging techniques' applications in LD research. We discuss holotomography and vibrational spectroscopic microscopy, emphasizing their potential for studying LDs without molecular labels, and we highlight the growing integration of artificial intelligence. Clinical applications in disease diagnosis and therapy are also considered.
Collapse
Affiliation(s)
- Hyeonwoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Seungeun Oh
- Department of Physics, Department of Cellular Molecular Medicine, University of California, San Diego, CA 2093, USA
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), Gwangju 61751, Republic of Korea; Department of Systems Biotechnology, Chung-Ang University Anseong-si, Gyeonggi-do 17546, Republic of Korea
| | - Kwang Suk Lee
- Department of Urology, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul 06229, Republic of Korea
| | - YongKeun Park
- Department of Physics, KAIST, Daejeon 34141, Republic of Korea; KAIST Institute for Health Science and Technology, KAIST, Daejeon 34141, Republic of Korea; Tomocube Inc., Daejeon 34109, Republic of Korea.
| |
Collapse
|
27
|
Cabodevilla AG, Son N, Goldberg IJ. Intracellular lipase and regulation of the lipid droplet. Curr Opin Lipidol 2024; 35:85-92. [PMID: 38447014 PMCID: PMC10919935 DOI: 10.1097/mol.0000000000000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
PURPOSE OF REVIEW Lipid droplets are increasingly recognized as distinct intracellular organelles that have functions exclusive to the storage of energetic lipids. Lipid droplets modulate macrophage inflammatory phenotype, control the availability of energy for muscle function, store excess lipid, sequester toxic lipids, modulate mitochondrial activity, and allow transfer of fatty acids between tissues. RECENT FINDINGS There have been several major advances in our understanding of the formation, dissolution, and function of this organelle during the past two years. These include new information on movement and partition of amphipathic proteins between the cytosol and lipid droplet surface, molecular determinants of lipid droplet formation, and pathways leading to lipid droplet hydrophobic lipid formation. Rapid advances in mitochondrial biology have also begun to define differences in their function and partnering with lipid droplets to modulate lipid storage versus oxidation. SUMMARY This relationship of lipid droplets biology and cellular function provides new understanding of an important cellular organelle that influences muscle function, adipose lipid storage, and diseases of lipotoxicity.
Collapse
Affiliation(s)
- Ainara G Cabodevilla
- Division of Endocrinology, New York University Grossman School of Medicine, New York, New York, USA
| | | | | |
Collapse
|
28
|
Mathiowetz AJ, Olzmann JA. Lipid droplets and cellular lipid flux. Nat Cell Biol 2024; 26:331-345. [PMID: 38454048 PMCID: PMC11228001 DOI: 10.1038/s41556-024-01364-4] [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] [Received: 07/30/2023] [Accepted: 01/24/2024] [Indexed: 03/09/2024]
Abstract
Lipid droplets are dynamic organelles that store neutral lipids, serve the metabolic needs of cells, and sequester lipids to prevent lipotoxicity and membrane damage. Here we review the current understanding of the mechanisms of lipid droplet biogenesis and turnover, the transfer of lipids and metabolites at membrane contact sites, and the role of lipid droplets in regulating fatty acid flux in lipotoxicity and cell death.
Collapse
Affiliation(s)
- Alyssa J Mathiowetz
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA.
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
29
|
Cinato M, Andersson L, Miljanovic A, Laudette M, Kunduzova O, Borén J, Levin MC. Role of Perilipins in Oxidative Stress-Implications for Cardiovascular Disease. Antioxidants (Basel) 2024; 13:209. [PMID: 38397807 PMCID: PMC10886189 DOI: 10.3390/antiox13020209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/12/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
Oxidative stress is the imbalance between the production of reactive oxygen species (ROS) and antioxidants in a cell. In the heart, oxidative stress may deteriorate calcium handling, cause arrhythmia, and enhance maladaptive cardiac remodeling by the induction of hypertrophic and apoptotic signaling pathways. Consequently, dysregulated ROS production and oxidative stress have been implicated in numerous cardiac diseases, including heart failure, cardiac ischemia-reperfusion injury, cardiac hypertrophy, and diabetic cardiomyopathy. Lipid droplets (LDs) are conserved intracellular organelles that enable the safe and stable storage of neutral lipids within the cytosol. LDs are coated with proteins, perilipins (Plins) being one of the most abundant. In this review, we will discuss the interplay between oxidative stress and Plins. Indeed, LDs and Plins are increasingly being recognized for playing a critical role beyond energy metabolism and lipid handling. Numerous reports suggest that an essential purpose of LD biogenesis is to alleviate cellular stress, such as oxidative stress. Given the yet unmet suitability of ROS as targets for the intervention of cardiovascular disease, the endogenous antioxidant capacity of Plins may be beneficial.
Collapse
Affiliation(s)
- Mathieu Cinato
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Linda Andersson
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Azra Miljanovic
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Marion Laudette
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University—Paul Sabatier, 31432 Toulouse, France;
| | - Jan Borén
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| | - Malin C. Levin
- Department of Molecular and Clinical Medicine/Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy, University of Gothenburg, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden; (M.C.); (L.A.); (A.M.); (M.L.); (J.B.)
| |
Collapse
|
30
|
Gao X, Jian L, Zhang L, Xu Y, Zhao Y, Yang Y, Yuan Y, Wang Y, Xu S, Ren B, Li Z, Wang C, Li J, Gu Y, Ye J. Perilipin 5 protects the mitochondrial oxidative functions and improves the alcoholic liver injury in mice. Liver Int 2024; 44:357-369. [PMID: 37933091 DOI: 10.1111/liv.15775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/20/2023] [Accepted: 10/20/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND AND AIMS Alcohol consumption is a well-established risk factor for the onset and progression of hepatic steatosis. Perilipin 5 (Plin5), a lipid droplet protein, is an important protective factor against hepatic lipotoxicity induced by excessive lipolysis, but its role and molecular mechanism in alcoholic liver disease (ALD) are not fully elucidated. METHODS The optimized National Institute on Alcohol Abuse and Alcoholism model was used to construct ALD model mice. Automatic biochemical analyser was used for Biochemical Parameters. The primary hepatocytes and Plin5-overexpressed HepG2 cells (including full-length Plin5 and Plin5 deleting 444-464 aa) were used for in vitro experiment. Haematoxylin and Eosin staining, Oil Red O staining, Bodipy 493/503 staining, Periodic Acid-Schiff staining, immunohistochemistry and JC-1 staining were used to evaluate cell morphology, lipids, glycogen, inflammation and membrane potential. Commercially kits are used to detect glycolipid metabolites, such as triglycerides, glycogen, glucose, reactive oxygen species, lactic acids, ketone bodies. Fluorescently labelled deoxyglucose, NBDG, was used for glucose intake. An XF96 extracellular flux analyser was used to determinate oxygen consumption rate in hepatocytes. The morphological and structural damage of mitochondria was evaluated by electron microscopy. Classical ultracentrifugation is used to separate the subcellular organelles of tissues and cells. Immunoblotting and qPCR were used to detect changes in mRNA and protein levels of related genes. RESULTS Our results showed that the expression of Plin5 in mouse livers was enhanced by alcohol intake, and Plin5 deficiency aggravated the alcohol-induced liver injury. To clarify the mechanism, we found that Plin5 deficiency significantly elevated the hepatic NADH levels and ketone body production in the alcohol-treated mice. As NADH elevation could promote the reduction of pyruvate into lactate and then inhibit the gluconeogenesis, alcohol-treated Plin5-deficient mice exhibited more lactate production and severer hypoglycemia. These results implied that Plin5 deficiency impaired the mitochondrial oxidative functions in the presence of alcohol. In addition, we demonstrated that Plin5 could be recruited onto mitochondria by alcohol, while Plin5 without mitochondrial targeting sequences lost its mitochondrial protection functions. CONCLUSION Collectively, this study demonstrated that the mitochondrial Plin5 could protect the alcohol-induced mitochondrial injury, which provides an important new insight on the roles of Plin5 in highly oxidative tissues.
Collapse
Affiliation(s)
- Xing Gao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Lele Jian
- State Key Laboratory of Cancer Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
- Shaanxi Provincial Corps, Chinese People's Armed Police Force, Xi'an, China
| | - Lijun Zhang
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuqiao Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuanlin Zhao
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Ying Yang
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuan Yuan
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yuying Wang
- State Key Laboratory of Cancer Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Shenhui Xu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bincheng Ren
- Department of Rheumatology and Immunology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Zimeng Li
- State Key Laboratory of Cancer Biology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Chao Wang
- Department of Pathology, The General Hospital of Western Theater Command, Chengdu, China
| | - Jing Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Yu Gu
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Jing Ye
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| |
Collapse
|
31
|
Chen W, Lu Y, Zhang Y, Wu J, McVicar A, Chen Y, Zhu S, Zhu G, Lu Y, Zhang J, McConnell M, Li YP. Cbfβ regulates Wnt/β-catenin, Hippo/Yap, and TGFβ signaling pathways in articular cartilage homeostasis and protects from ACLT surgery-induced osteoarthritis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575763. [PMID: 38293189 PMCID: PMC10827176 DOI: 10.1101/2024.01.15.575763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
As the most common degenerative joint disease, osteoarthritis (OA) contributes significantly to pain and disability during aging. Several genes of interest involved in articular cartilage damage in OA have been identified. However, the direct causes of OA are poorly understood. Evaluating the public human RNA-seq dataset showed that Cbfβ, (subunit of a heterodimeric Cbfβ/Runx1,Runx2, or Runx3 complex) expression is decreased in the cartilage of patients with OA. Here, we found that the chondrocyte-specific deletion of Cbfβ in tamoxifen-induced Cbfβf/fCol2α1-CreERT mice caused a spontaneous OA phenotype, worn articular cartilage, increased inflammation, and osteophytes. RNA-sequencing analysis showed that Cbfβ deficiency in articular cartilage resulted in reduced cartilage regeneration, increased canonical Wnt signaling and inflammatory response, and decreased Hippo/YAP signaling and TGF-β signaling. Immunostaining and western blot validated these RNA-seq analysis results. ACLT surgery-induced OA decreased Cbfβ and Yap expression and increased active β-catenin expression in articular cartilage, while local AAV-mediated Cbfβ overexpression promoted Yap expression and diminished active β-catenin expression in OA lesions. Remarkably, AAV-mediated Cbfβ overexpression in knee joints of mice with OA showed the significant protective effect of Cbfβ on articular cartilage in the ACLT OA mouse model. Overall, this study, using loss-of-function and gain-of-function approaches, uncovered that low expression of Cbfβ may be the cause of OA. Moreover, Local admission of Cbfβ may rescue and protect OA through decreasing Wnt/β-catenin signaling, and increasing Hippo/Yap signaling and TGFβ/Smad2/3 signaling in OA articular cartilage, indicating that local Cbfβ overexpression could be an effective strategy for treatment of OA.
Collapse
Affiliation(s)
- Wei Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yun Lu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yan Zhang
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jinjin Wu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Abigail McVicar
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yilin Chen
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Siyu Zhu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Guochun Zhu
- Department of Pathology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - You Lu
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Jiayang Zhang
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Matthew McConnell
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| | - Yi-Ping Li
- Division in Cellular and Molecular Medicine, Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
32
|
Miner GE, Smith SY, Showalter WK, So CM, Ragusa JV, Powers AE, Zanellati MC, Hsu CH, Marchan MF, Cohen S. Contact-FP: A Dimerization-Dependent Fluorescent Protein Toolkit for Visualizing Membrane Contact Site Dynamics. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241228911. [PMID: 38327561 PMCID: PMC10846013 DOI: 10.1177/25152564241228911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 01/11/2024] [Indexed: 02/09/2024]
Abstract
Membrane contact sites (MCSs) are sites of close apposition between two organelles used to exchange ions, lipids, and information. Cells respond to changing environmental or developmental conditions by modulating the number, extent, or duration of MCSs. Because of their small size and dynamic nature, tools to study the dynamics of MCSs in live cells have been limited. Dimerization-dependent fluorescent proteins (ddFPs) targeted to organelle membranes are an ideal tool for studying MCS dynamics because they reversibly interact to fluoresce specifically at the interface between two organelles. Here, we build on previous work using ddFPs as sensors to visualize the morphology and dynamics of MCSs. We engineered a suite of ddFPs called Contact-FP that targets ddFP monomers to lipid droplets (LDs), the endoplasmic reticulum (ER), mitochondria, peroxisomes, lysosomes, plasma membrane, caveolae, and the cytoplasm. We show that these probes correctly localize to their target organelles. Using LDs as a test case, we demonstrate that Contact-FP pairs specifically localize to the interface between two target organelles. Titration of LD-mitochondria ddFPs revealed that these sensors can be used at high concentrations to drive MCSs or can be titrated down to minimally perturb and visualize endogenous MCSs. We show that Contact-FP probes can be used to: (1) visualize LD-mitochondria MCS dynamics, (2) observe changes in LD-mitochondria MCS dynamics upon overexpression of PLIN5, a known LD-mitochondrial tether, and (3) visualize two MCSs that share one organelle simultaneously (e.g., LD-mitochondria and LD-ER MCSs). Contact-FP probes can be optimized to visualize MCSs between any pair of organelles represented in the toolkit.
Collapse
Affiliation(s)
- Gregory E. Miner
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sidney Y. Smith
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Wendy K. Showalter
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Christina M. So
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Joey V. Ragusa
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Alex E. Powers
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Maria Clara Zanellati
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Chih-Hsuan Hsu
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michelle F. Marchan
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Sarah Cohen
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
33
|
Li H, Seessle J, Staffer S, Tuma-Kellner S, Poschet G, Herrmann T, Chamulitrat W. FATP4 deletion in liver cells induces elevation of extracellular lipids via metabolic channeling towards triglycerides and lipolysis. Biochem Biophys Res Commun 2023; 687:149161. [PMID: 37931418 DOI: 10.1016/j.bbrc.2023.149161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/13/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
Evidence from mice with global deletion of fatty-acid transport protein4 (FATP4) indicates its role on β-oxidation and triglycerides (TG) metabolism. We reported that plasma glycerol and free fatty acids (FA) were increased in liver-specific Fatp4 deficient (L-FATP4-/-) mice under dietary stress. We hypothesized that FATP4 may mediate hepatocellular TG lipolysis. Here, we demonstrated that L-FATP4-/- mice showed an increase in these blood lipids, liver TG, and subcutaneous fat weights. We therefore studied TG metabolism in response to oleate treatment in two experimental models using FATP4-knockout HepG2 (HepKO) cells and L-FATP4-/- hepatocytes. Both FATP4-deificient liver cells showed a significant decrease in β-oxidation products by ∼30-35% concomitant with marked upregulation of CD36, FATP2, and FATP5 as well as lipoprotein microsomal-triglyceride-transfer protein genes. By using 13C3D5-glycerol, HepKO cells displayed an increase in metabolically labelled TG species which were further increased with oleate treatment. This increase was concomitant with a step-wise elevation of TG in cells and supernatants as well as the secretion of cholesterol very low-density and high-density lipoproteins. Upon analyzing TG lipolytic enzymes, both mutant liver cells showed marked upregulated expression of hepatic lipase, while that of hormone-sensitive lipase and adipose-triglyceride lipase was downregulated. Lipolysis measured by extracellular glycerol and free FA was indeed increased in mutant cells, and this event was exacerbated by oleate treatment. Taken together, FATP4 deficiency in liver cells led to a metabolic shift from β-oxidation towards lipolysis-directed TG and lipoprotein secretion, which is in line with an association of FATP4 polymorphisms with blood lipids.
Collapse
Affiliation(s)
- Huili Li
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany; Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, Hubei, China
| | - Jessica Seessle
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Simone Staffer
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Sabine Tuma-Kellner
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany
| | - Gernot Poschet
- Metabolomics Core Technology Platform, Centre for Organismal Studies, University of Heidelberg, 69120, Heidelberg, Germany
| | - Thomas Herrmann
- Westkuesten Hospital, Esmarchstraße 50, 25746, Heide, Germany
| | - Walee Chamulitrat
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
34
|
Li X, Gamuyao R, Wu ML, Cho WJ, Kurtz NB, King SV, Petersen R, Stabley DR, Lindow C, Climer L, Shirinifard A, Ferrara F, Throm RE, Robinson CG, Carisey A, Tebo AG, Chang CL. A fluorogenic complementation tool kit for interrogating lipid droplet-organelle interaction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569289. [PMID: 38076863 PMCID: PMC10705429 DOI: 10.1101/2023.11.29.569289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Contact sites between lipid droplets and other organelles are essential for cellular lipid and energy homeostasis. Detection of these contact sites at nanometer scale over time in living cells is challenging. Here, we developed a tool kit for detecting contact sites based on Fluorogen-Activated Bimolecular complementation at CONtact sites, FABCON, using a reversible, low affinity split fluorescent protein, splitFAST. FABCON labels contact sites with minimal perturbation to organelle interaction. Via FABCON, we quantitatively demonstrated that endoplasmic reticulum (ER)- and mitochondria (mito)-lipid droplet contact sites are dynamic foci in distinct metabolic conditions, such as during lipid droplet biogenesis and consumption. An automated analysis pipeline further classified individual contact sites into distinct subgroups based on size, likely reflecting differential regulation and function. Moreover, FABCON is generalizable to visualize a repertoire of organelle contact sites including ER-mito. Altogether, FABCON reveals insights into the dynamic regulation of lipid droplet-organelle contact sites and generates new hypotheses for further mechanistical interrogation during metabolic switch.
Collapse
Affiliation(s)
- Xiao Li
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Rico Gamuyao
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Ming-Lun Wu
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Woo Jung Cho
- Cell and Tissue Imaging Center, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Nathan B. Kurtz
- Cell and Tissue Imaging Center, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Sharon V. King
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - R.A. Petersen
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Daniel R. Stabley
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Caleb Lindow
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Leslie Climer
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Francesca Ferrara
- Vector Production and Development Laboratory, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Robert E. Throm
- Vector Production and Development Laboratory, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Camenzind G. Robinson
- Cell and Tissue Imaging Center, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Alex Carisey
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Alison G. Tebo
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, United States
| | - Chi-Lun Chang
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| |
Collapse
|