1
|
Kovaleva T, Gainullin M, Mukhina I, Pershin V, Matskova L. Cofilin(s) and Mitochondria: Function Beyond Actin Dynamics. Int J Mol Sci 2025; 26:4094. [PMID: 40362336 PMCID: PMC12071280 DOI: 10.3390/ijms26094094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/18/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
ADF/cofilins form a family of small, widely expressed actin-binding proteins, regulating actin dynamics in various cellular and physiological processes in all eukaryotes, from yeasts to animals. Changes in the expression of the ADF/cofilin family proteins have been demonstrated under various pathological conditions. The well-established role of cofilin in migration, invasion, epithelial-mesenchymal transition, apoptosis, resistance to radiotherapy and chemotherapy, immune escape, and transcriptional dysregulation in malignant tumors is primarily attributed to its actin-modifying activity. Moreover, drugs targeting this function of cofilin have been developed for cancer treatment. However, its multilevel regulation, highly diverse effects across various pathological conditions, and conflicting data on the functional consequences of altered cofilin expression have prompted us to explore additional roles of cofilin-beyond actin modulation-particularly its involvement in lipid metabolism and mitochondrial homeostasis. Here, we review recent data on the expression of ADF/cofilin family proteins in various pathologies, account for the mutations and post-translational modifications of these proteins and their functional consequences, dwell on the role of K63-type ubiquitination of cofilin for its involvement in lipid metabolism and mitochondrial homeostasis, more specifically, a process of mitochondrial division or mitofission, point out conflicting data in cofilin research, and describe prospects for future studies of cofilin functions.
Collapse
Affiliation(s)
- Tatiana Kovaleva
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin Sq., 603005 Nizhny Novgorod, Russia; (I.M.); (V.P.)
| | | | - Irina Mukhina
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin Sq., 603005 Nizhny Novgorod, Russia; (I.M.); (V.P.)
| | - Vladimir Pershin
- Institute of Fundamental Medicine, Privolzhsky Research Medical University, 10/1 Minin Sq., 603005 Nizhny Novgorod, Russia; (I.M.); (V.P.)
| | - Liudmila Matskova
- Microbiology and Tumor Biology Center (MTC), Karolinska Institutet, Solnavägen 9, Q8C, 17165 Stockholm, Sweden
- Institute of Molecular Biology and Biophysics, Federal Research Center of Fundamental and Translational Medicine (IMBB FRC FTM), 2/12, Timakova Street, 630117 Novosibirsk, Russia
| |
Collapse
|
2
|
You D, Kang S. JMJD8 Regulates Adipocyte Hypertrophy Through the Interaction With Perilipin 2. Diabetes 2025; 74:458-471. [PMID: 39787420 PMCID: PMC11926275 DOI: 10.2337/db23-0883] [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: 11/05/2023] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
ARTICLE HIGHLIGHTS New research builds on previous findings that JMJD8 mediates insulin resistance by promoting adipocyte hypertrophy. We identified PLIN2 as a binding partner of JMJD8 using proteomics approaches. This study reveals a physical interaction between JMJD8 and PLIN2, which plays a crucial role in driving adipocyte hypertrophy and insulin resistance. JMJD8 suppresses fasting-induced lipophagy and reduces energy production by inhibiting PLIN2 phosphorylation. These findings highlight the importance of JMJD8 and PLIN2 in regulating lipid droplet homeostasis and suggest a potential mechanism for controlling fat mobilization during energy deprivation.
Collapse
Affiliation(s)
- Dongjoo You
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, CA
| | - Sona Kang
- Nutritional Sciences and Toxicology Department, University of California, Berkeley, Berkeley, CA
| |
Collapse
|
3
|
Tiwari P, Brooks D, Geisbrecht ER. Overexpression of Drosophila NUAK or Constitutively-Active Formin-Like Promotes the Formation of Aberrant Myofibrils. Cytoskeleton (Hoboken) 2025. [PMID: 39876757 DOI: 10.1002/cm.21999] [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: 10/03/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025]
Abstract
Muscle development and maintenance is central to the normal functioning of animals. Muscle tissues exhibit high levels of activity and require the dynamic turnover of proteins. An actomyosin scaffold functions with additional proteins comprising the basic contractile subunit of striated muscle, known as the sarcomere. Drosophila muscles are similar to vertebrate muscles in composition and they share a similar mechanism of development. Drosophila NUAK (NUAK) is the homolog of NUAK1 and NUAK2 in vertebrates. NUAK belongs to the family of AMP-activated protein kinases (AMPKs), a group of proteins with broad and overlapping cellular targets. Here we confirm that NUAK dynamically modulates larval muscle sarcomere size as upregulation of NUAK produces longer sarcomeres, including increased thin filament lengths. Furthermore, NUAK overexpression results in aberrant myofibers above the nuclei plane, upregulation of Formin-like (Frl), and an increase in newly synthesized proteins at sites consistent with actin filament assembly. Expression of constitutively-active Frl also produces aberrant myofibers similar to NUAK overexpression. These results taken together strongly suggest a functional link between NUAK and Frl in myofibril formation in an in vivo setting.
Collapse
Affiliation(s)
- Prabhat Tiwari
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas, USA
| | - David Brooks
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas, USA
| | - Erika R Geisbrecht
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
4
|
Zhang W, Tian X, Zhu Y, Yang L, Li Z, Yu M. An intelligent NIR fluorescent dye with dual response to pH and viscosity for investigating the interactions between LDs and mitochondria. Talanta 2025; 281:126849. [PMID: 39276568 DOI: 10.1016/j.talanta.2024.126849] [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: 07/02/2024] [Revised: 08/19/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
The interaction between lipid droplets and mitochondria plays a pivotal role in biological processes including cellular stress, metabolic homeostasis, cellular autophagy and apoptosis. Deciphering the complex interplay between lipid droplets and mitochondria is essential for gaining insights into the fundamental workings of the cell and can have broad implications for the development of therapeutic strategies for various diseases, including metabolic disorders, neurodegenerative diseases, and cancer. In this study, we develop a pH and viscosity-responsive near-infrared (NIR) fluorescent probe PTOH to investigate the interaction between lipid droplets and mitochondria. This probe demonstrates a significant enhancement in fluorescence intensity at 470 nm when the pH increases, while under acidic conditions, its fluorescence intensity at 730 nm intensifies by a factor of 35 with rising system viscosity. Cell imaging experiments revealed that PTOH can effectively discriminate between normal and cancerous cells, as well as detect intracellular pH and viscosity alterations induced by drugs. Additionally, PTOH is utilized to visualize the interaction between lipid droplets and mitochondria and to differentiate between cellular autophagy and apoptosis phenomena, providing a valuable tool for elucidating the mechanisms underlying lipid droplet-mitochondria interactions and their associated diseases.
Collapse
Affiliation(s)
- Wei Zhang
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Xing Tian
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China
| | - Yanxi Zhu
- Linyi Key Laboratory of Nanomedicine, Linyi People's Hospital, Linyi, 276000, China.
| | - Lei Yang
- Shandong Provincial Key Laboratory of Detection Technology for Tumor Markers, College of Chemistry and Chemical Engineering, Linyi University, Linyi, 276000, China
| | - Zhanxian Li
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China.
| | - Mingming Yu
- College of Chemistry, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
5
|
Chen L, Hao J, Zhang J, Wu J, Ren Z. Rosiglitazone-induced white adipocyte browning is regulated by actin and Myh9. Life Sci 2024; 359:123217. [PMID: 39510170 DOI: 10.1016/j.lfs.2024.123217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 10/22/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024]
Abstract
AIMS This study investigates the role of actin polymerization and Myh9 in mediating lipid droplet (LD) fission during rosiglitazone-induced browning of white adipocytes. The aim is to understand how LD splitting might contribute to the beige conversion of white adipose tissue, providing insights into adipocyte plasticity and metabolic regulation. MATERIALS AND METHODS C3H10 T1/2-differentiated adipocytes were used as a classical model to study white adipocyte browning. Rosiglitazone was applied to induce browning, and the interactions between LDs and actin, as well as the distribution of Myh9, were assessed using immunofluorescence and Western blotting. In vivo, we employed a microfilament inhibitor to block actin polymerization in cold-stimulated mice and evaluated changes in LD morphology and browning. Furthermore, dynamic live-cell imaging using confocal microscopy was conducted to observe the real-time behavior of LDs during the browning process and to determine whether they undergo fission. MAIN FINDINGS Our results demonstrate that rosiglitazone significantly induces LD size reduction, a process correlated with the increased contact of LDs with microfilaments. Inhibition of actin polymerization prevented both the reduction in LD size and the browning of white adipocytes, indicating that actin plays a critical role. Myh9 was enriched at the LD fission sites, forming a structure resembling a contractile ring. Overexpression of Myh9 promoted the shrinkage of LD, suggesting that it may be involved in LD fission. SIGNIFICANCE This study identifies actin and Myh9 as key regulators of LD fission in rosiglitazone-induced browning of white adipocytes, offering new insights into the cellular mechanisms of adipocyte plasticity. The findings propose a novel pathway by which LD dynamics contribute to the beige conversion of white fat, with potential implications for metabolic disease therapies targeting adipocyte function and energy expenditure.
Collapse
Affiliation(s)
- Lupeng Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jingjie Hao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Junzhi Zhang
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Frontiers Science Center for Animal Breeding and Sustainable Production, Wuhan, Hubei 430070, China; Hubei Hongshan Laboratory, Wuhan, Hubei 430070, China.
| |
Collapse
|
6
|
Jia QH, Cao YZ, Xing YX, Guan HB, Ma CL, Li X, Tian WH, Li ZJ, Tian YD, Li GX, Jiang RR, Kang XT, Liu XJ, Li H. LncRNA lncLLM Facilitates Lipid Deposition by Promoting the Ubiquitination of MYH9 in Chicken LMH Cells. Int J Mol Sci 2024; 25:10316. [PMID: 39408647 PMCID: PMC11477197 DOI: 10.3390/ijms251910316] [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: 08/22/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
The liver plays an important role in regulating lipid metabolism in animals. This study investigated the function and mechanism of lncLLM in liver lipid metabolism in hens at the peak of egg production. The effect of lncLLM on intracellular lipid content in LMH cells was evaluated by qPCR, Oil Red O staining, and detection of triglyceride (TG) and cholesterol (TC) content. The interaction between lncLLM and MYH9 was confirmed by RNA purification chromatin fractionation (CHIRP) and RNA immunoprecipitation (RIP) analysis. The results showed that lncLLM increased the intracellular content of TG and TC and promoted the expression of genes related to lipid synthesis. It was further found that lncLLM had a negative regulatory effect on the expression level of MYH9 protein in LMH cells. The intracellular TG and TC content of MYH9 knockdown cells increased, and the expression of genes related to lipid decomposition was significantly reduced. In addition, this study confirmed that the role of lncLLM is at least partly through mediating the ubiquitination of MYH9 protein to accelerate the degradation of MYH9 protein. This discovery provides a new molecular target for improving egg-laying performance in hens and treating fatty liver disease in humans.
Collapse
Affiliation(s)
- Qi-Hui Jia
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
| | - Yu-Zhu Cao
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
| | - Yu-Xin Xing
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
| | - Hong-Bo Guan
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
| | - Cheng-Lin Ma
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
| | - Xin Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
| | - Wei-Hua Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
| | - Zhuan-Jian Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450046, China
| | - Ya-Dong Tian
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450046, China
| | - Guo-Xi Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450046, China
| | - Rui-Rui Jiang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450046, China
| | - Xiang-Tao Kang
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450046, China
| | - Xiao-Jun Liu
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450046, China
| | - Hong Li
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou 450046, China; (Q.-H.J.); (Y.-Z.C.); (Y.-X.X.); (H.-B.G.); (C.-L.M.); (X.L.); (W.-H.T.); (Z.-J.L.); (Y.-D.T.); (G.-X.L.); (R.-R.J.); (X.-T.K.)
- Key Laboratory of Livestock and Poultry Resources (Poultry) Evaluation and Utilization, Ministry of Agriculture and Rural Affairs, Henan Agricultural University, Zhengzhou 450046, China
- International Joint Research Laboratory for Poultry Breeding of Henan, Henan Agricultural University, Zhengzhou 450046, China
| |
Collapse
|
7
|
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
|
8
|
Omata Y, Sato R, Mishiro-Sato E, Kano K, Ueda H, Hara-Nishimura I, Shimada TL. Lipid droplets in Arabidopsis thaliana leaves contain myosin-binding proteins and enzymes associated with furan-containing fatty acid biosynthesis. FRONTIERS IN PLANT SCIENCE 2024; 15:1331479. [PMID: 38495375 PMCID: PMC10940516 DOI: 10.3389/fpls.2024.1331479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/13/2024] [Indexed: 03/19/2024]
Abstract
Lipid droplets (LDs) are lipid storage organelles in plant leaves and seeds. Seed LD proteins are well known, and their functions in lipid metabolism have been characterized; however, many leaf LD proteins remain to be identified. We therefore isolated LDs from leaves of the leaf LD-overaccumulating mutant high sterol ester 1 (hise1) of Arabidopsis thaliana by centrifugation or co-immunoprecipitation. We then performed LD proteomics by mass spectrometry and identified 3,206 candidate leaf LD proteins. In this study, we selected 31 candidate proteins for transient expression assays using a construct encoding the candidate protein fused with green fluorescent protein (GFP). Fluorescence microscopy showed that MYOSIN BINDING PROTEIN14 (MYOB14) and two uncharacterized proteins localized to LDs labeled with the LD marker. Subcellular localization analysis of MYOB family members revealed that MYOB1, MYOB2, MYOB3, and MYOB5 localized to LDs. LDs moved along actin filaments together with the endoplasmic reticulum. Co-immunoprecipitation of myosin XIK with MYOB2-GFP or MYOB14-GFP suggested that LD-localized MYOBs are involved in association with the myosin XIK-LDs. The two uncharacterized proteins were highly similar to enzymes for furan fatty acid biosynthesis in the photosynthetic bacterium Cereibacter sphaeroides, suggesting a relationship between LDs and furan fatty acid biosynthesis. Our findings thus reveal potential molecular functions of LDs and provide a valuable resource for further studies of the leaf LD proteome.
Collapse
Affiliation(s)
- Yuto Omata
- Faculty of Horticulture, Chiba University, Matsudo, Japan
| | - Reina Sato
- Faculty of Horticulture, Chiba University, Matsudo, Japan
| | - Emi Mishiro-Sato
- World Premier International Research Center Initiative-Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
| | - Keiko Kano
- World Premier International Research Center Initiative-Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Nagoya, Japan
| | - Haruko Ueda
- Faculty of Science and Engineering, Konan University, Kobe, Japan
| | | | - Takashi L. Shimada
- Faculty of Horticulture, Chiba University, Matsudo, Japan
- Graduate School of Horticulture, Chiba University, Matsudo, Japan
- Plant Molecular Science Center, Chiba University, Chiba, Japan
- Research Center for Space Agriculture and Horticulture, Chiba University, Matsudo, Japan
| |
Collapse
|
9
|
Giedt MS, Thomalla JM, White RP, Johnson MR, Lai ZW, Tootle TL, Welte MA. Adipose triglyceride lipase promotes prostaglandin-dependent actin remodeling by regulating substrate release from lipid droplets. Development 2023; 150:dev201516. [PMID: 37306387 PMCID: PMC10281261 DOI: 10.1242/dev.201516] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
Lipid droplets (LDs), crucial regulators of lipid metabolism, accumulate during oocyte development. However, their roles in fertility remain largely unknown. During Drosophila oogenesis, LD accumulation coincides with the actin remodeling necessary for follicle development. Loss of the LD-associated Adipose Triglyceride Lipase (ATGL) disrupts both actin bundle formation and cortical actin integrity, an unusual phenotype also seen when the prostaglandin (PG) synthase Pxt is missing. Dominant genetic interactions and PG treatment of follicles indicate that ATGL acts upstream of Pxt to regulate actin remodeling. Our data suggest that ATGL releases arachidonic acid (AA) from LDs to serve as the substrate for PG synthesis. Lipidomic analysis detects AA-containing triglycerides in ovaries, and these are increased when ATGL is lost. High levels of exogenous AA block follicle development; this is enhanced by impairing LD formation and suppressed by reducing ATGL. Together, these data support the model that AA stored in LD triglycerides is released by ATGL to drive the production of PGs, which promote the actin remodeling necessary for follicle development. We speculate that this pathway is conserved across organisms to regulate oocyte development and promote fertility.
Collapse
Affiliation(s)
- Michelle S. Giedt
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Roger P. White
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew R. Johnson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Zon Weng Lai
- Harvard T.H. Chan Advanced Multi-omics Platform, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tina L. Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Michael A. Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
10
|
Vitriol EA, Quintanilla MA, Tidei JJ, Troughton LD, Cody A, Cisterna BA, Jane ML, Oakes PW, Beach JR. Nonmuscle myosin 2 filaments are processive in cells. Biophys J 2023; 122:3678-3689. [PMID: 37218133 PMCID: PMC10541485 DOI: 10.1016/j.bpj.2023.05.014] [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: 02/24/2023] [Revised: 04/26/2023] [Accepted: 05/18/2023] [Indexed: 05/24/2023] Open
Abstract
Directed transport of cellular components is often dependent on the processive movements of cytoskeletal motors. Myosin 2 motors predominantly engage actin filaments of opposing orientation to drive contractile events and are therefore not traditionally viewed as processive. However, recent in vitro experiments with purified nonmuscle myosin 2 (NM2) demonstrated myosin 2 filaments could move processively. Here, we establish processivity as a cellular property of NM2. Processive runs in central nervous system-derived CAD cells are most apparent on bundled actin in protrusions that terminate at the leading edge. We find that processive velocities in vivo are consistent with in vitro measurements. NM2 makes these processive runs in its filamentous form against lamellipodia retrograde flow, though anterograde movement can still occur in the absence of actin dynamics. Comparing the processivity of NM2 isoforms, we find that NM2A moves slightly faster than NM2B. Finally, we demonstrate that this is not a cell-specific property, as we observe processive-like movements of NM2 in the lamella and subnuclear stress fibers of fibroblasts. Collectively, these observations further broaden NM2 functionality and the biological processes in which the already ubiquitous motor can contribute.
Collapse
Affiliation(s)
- Eric A Vitriol
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia.
| | - Melissa A Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Joseph J Tidei
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Lee D Troughton
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Abigail Cody
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois
| | - Bruno A Cisterna
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia
| | - Makenzie L Jane
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia
| | - Patrick W Oakes
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois.
| | - Jordan R Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois.
| |
Collapse
|
11
|
Davis J, Meyer T, Smolnig M, Smethurst DG, Neuhaus L, Heyden J, Broeskamp F, Edrich ES, Knittelfelder O, Kolb D, Haar TVD, Gourlay CW, Rockenfeller P. A dynamic actin cytoskeleton is required to prevent constitutive VDAC-dependent MAPK signalling and aberrant lipid homeostasis. iScience 2023; 26:107539. [PMID: 37636069 PMCID: PMC10450525 DOI: 10.1016/j.isci.2023.107539] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/14/2023] [Accepted: 07/28/2023] [Indexed: 08/29/2023] Open
Abstract
The dynamic nature of the actin cytoskeleton is required to coordinate many cellular processes, and a loss of its plasticity has been linked to accelerated cell aging and attenuation of adaptive response mechanisms. Cofilin is an actin-binding protein that controls actin dynamics and has been linked to mitochondrial signaling pathways that control drug resistance and cell death. Here we show that cofilin-driven chronic depolarization of the actin cytoskeleton activates cell wall integrity mitogen-activated protein kinase (MAPK) signalling and disrupts lipid homeostasis in a voltage-dependent anion channel (VDAC)-dependent manner. Expression of the cof1-5 mutation, which reduces the dynamic nature of actin, triggers loss of cell wall integrity, vacuole fragmentation, disruption of lipid homeostasis, lipid droplet (LD) accumulation, and the promotion of cell death. The integrity of the actin cytoskeleton is therefore essential to maintain the fidelity of MAPK signaling, lipid homeostasis, and cell health in S. cerevisiae.
Collapse
Affiliation(s)
- Jack Davis
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Thorsten Meyer
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| | - Martin Smolnig
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| | | | - Lisa Neuhaus
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| | - Jonas Heyden
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| | - Filomena Broeskamp
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| | | | - Oskar Knittelfelder
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Dagmar Kolb
- Medical University of Graz, Core Facility Ultrastructure Analysis, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Cell Biology, Histology and Embryology, Medical University of Graz, Neue Stiftingtalstraße 6/II, 8010 Graz, Austria
| | - Tobias von der Haar
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Campbell W. Gourlay
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Patrick Rockenfeller
- Chair of Biochemistry and Molecular Medicine, Center for Biomedical Education and Research (ZBAF), University of Witten/Herdecke (UW/H), Stockumer Str. 10, 58453 Witten, Germany
| |
Collapse
|
12
|
Ivanovska IL, Tobin MP, Bai T, Dooling LJ, Discher DE. Small lipid droplets are rigid enough to indent a nucleus, dilute the lamina, and cause rupture. J Cell Biol 2023; 222:e202208123. [PMID: 37212777 PMCID: PMC10202833 DOI: 10.1083/jcb.202208123] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/24/2023] [Accepted: 05/04/2023] [Indexed: 05/23/2023] Open
Abstract
The nucleus in many cell types is a stiff organelle, but fat-filled lipid droplets (FDs) in cytoplasm are seen to indent and displace the nucleus. FDs are phase-separated liquids with a poorly understood interfacial tension γ that determines how FDs interact with other organelles. Here, micron-sized FDs remain spherical as they indent peri-nuclear actomyosin and the nucleus, while causing local dilution of Lamin-B1 independent of Lamin-A,C and sometimes triggering nuclear rupture. Focal accumulation of the cytosolic DNA sensor cGAS at the rupture site is accompanied by sustained mislocalization of DNA repair factors to cytoplasm, increased DNA damage, and delayed cell cycle. Macrophages show FDs and engulfed rigid beads cause similar indentation dilution. Spherical shapes of small FDs indicate a high γ, which we measure for FDs mechanically isolated from fresh adipose tissue as ∼40 mN/m. This value is far higher than that of protein condensates, but typical of oils in water and sufficiently rigid to perturb cell structures including nuclei.
Collapse
Affiliation(s)
- Irena L. Ivanovska
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Tobin
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Tianyi Bai
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Lawrence J. Dooling
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E. Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
13
|
Hüsler D, Stauffer P, Hilbi H. Tapping lipid droplets: A rich fat diet of intracellular bacterial pathogens. Mol Microbiol 2023; 120:194-209. [PMID: 37429596 DOI: 10.1111/mmi.15120] [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: 05/03/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/12/2023]
Abstract
Lipid droplets (LDs) are dynamic and versatile organelles present in most eukaryotic cells. LDs consist of a hydrophobic core of neutral lipids, a phospholipid monolayer coat, and a variety of associated proteins. LDs are formed at the endoplasmic reticulum and have diverse roles in lipid storage, energy metabolism, membrane trafficking, and cellular signaling. In addition to their physiological cellular functions, LDs have been implicated in the pathogenesis of several diseases, including metabolic disorders, cancer, and infections. A number of intracellular bacterial pathogens modulate and/or interact with LDs during host cell infection. Members of the genera Mycobacterium, Legionella, Coxiella, Chlamydia, and Salmonella exploit LDs as a source of intracellular nutrients and membrane components to establish their distinct intracellular replicative niches. In this review, we focus on the biogenesis, interactions, and functions of LDs, as well as on their role in lipid metabolism of intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Dario Hüsler
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Pia Stauffer
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| | - Hubert Hilbi
- Institute of Medical Microbiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Han L, Huang D, Wu S, Liu S, Wang C, Sheng Y, Lu X, Broxmeyer HE, Wan J, Yang L. Lipid droplet-associated lncRNA LIPTER preserves cardiac lipid metabolism. Nat Cell Biol 2023; 25:1033-1046. [PMID: 37264180 PMCID: PMC10344779 DOI: 10.1038/s41556-023-01162-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 05/03/2023] [Indexed: 06/03/2023]
Abstract
Lipid droplets (LDs) are cellular organelles critical for lipid homeostasis, with intramyocyte LD accumulation implicated in metabolic disorder-associated heart diseases. Here we identify a human long non-coding RNA, Lipid-Droplet Transporter (LIPTER), essential for LD transport in human cardiomyocytes. LIPTER binds phosphatidic acid and phosphatidylinositol 4-phosphate on LD surface membranes and the MYH10 protein, connecting LDs to the MYH10-ACTIN cytoskeleton and facilitating LD transport. LIPTER and MYH10 deficiencies impair LD trafficking, mitochondrial function and survival of human induced pluripotent stem cell-derived cardiomyocytes. Conditional Myh10 deletion in mouse cardiomyocytes leads to LD accumulation, reduced fatty acid oxidation and compromised cardiac function. We identify NKX2.5 as the primary regulator of cardiomyocyte-specific LIPTER transcription. Notably, LIPTER transgenic expression mitigates cardiac lipotoxicity, preserves cardiac function and alleviates cardiomyopathies in high-fat-diet-fed and Leprdb/db mice. Our findings unveil a molecular connector role of LIPTER in intramyocyte LD transport, crucial for lipid metabolism of the human heart, and hold significant clinical implications for treating metabolic syndrome-associated heart diseases.
Collapse
Affiliation(s)
- Lei Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Dayang Huang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shiyong Wu
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sheng Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cheng Wang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yi Sheng
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiongbin Lu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jun Wan
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lei Yang
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
15
|
Amarasinghe I, Phillips W, Hill AF, Cheng L, Helbig KJ, Willms E, Monson EA. Cellular communication through extracellular vesicles and lipid droplets. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e77. [PMID: 38938415 PMCID: PMC11080893 DOI: 10.1002/jex2.77] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/06/2023] [Accepted: 02/15/2023] [Indexed: 06/29/2024]
Abstract
Cellular communication is essential for effective coordination of biological processes. One major form of intercellular communication occurs via the release of extracellular vesicles (EVs). These vesicles mediate intercellular communication through the transfer of their cargo and are actively explored for their role in various diseases and their potential therapeutic and diagnostic applications. Conversely, lipid droplets (LDs) are vesicles that transfer cargo within cells. Lipid droplets play roles in various diseases and evidence for their ability to transfer cargo between cells is emerging. To date, there has been little interdisciplinary research looking at the similarities and interactions between these two classes of small lipid vesicles. This review will compare the commonalities and differences between EVs and LDs including their biogenesis and secretion, isolation and characterisation methodologies, composition, and general heterogeneity and discuss challenges and opportunities in both fields.
Collapse
Affiliation(s)
- Irumi Amarasinghe
- School of Agriculture, Biomedicine and EnvironmentLa Trobe UniversityMelbourneAustralia
| | - William Phillips
- School of Agriculture, Biomedicine and EnvironmentLa Trobe UniversityMelbourneAustralia
- La Trobe Institute for Molecular SciencesLa Trobe UniversityMelbourneAustralia
| | - Andrew F. Hill
- Institute for Health and SportVictoria UniversityFootscrayVictoriaAustralia
| | - Lesley Cheng
- School of Agriculture, Biomedicine and EnvironmentLa Trobe UniversityMelbourneAustralia
- La Trobe Institute for Molecular SciencesLa Trobe UniversityMelbourneAustralia
| | - Karla J. Helbig
- School of Agriculture, Biomedicine and EnvironmentLa Trobe UniversityMelbourneAustralia
| | - Eduard Willms
- School of Agriculture, Biomedicine and EnvironmentLa Trobe UniversityMelbourneAustralia
- La Trobe Institute for Molecular SciencesLa Trobe UniversityMelbourneAustralia
| | - Ebony A. Monson
- School of Agriculture, Biomedicine and EnvironmentLa Trobe UniversityMelbourneAustralia
| |
Collapse
|
16
|
Vitriol EA, Quintanilla MA, Tidei JJ, Troughton LD, Cody A, Cisterna BA, Jane ML, Oakes PW, Beach JR. Non-muscle myosin 2 filaments are processive in cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.24.529920. [PMID: 36865321 PMCID: PMC9980172 DOI: 10.1101/2023.02.24.529920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
Directed transport of cellular components is often dependent on the processive movements of cytoskeletal motors. Myosin 2 motors predominantly engage actin filaments of opposing orientation to drive contractile events, and are therefore not traditionally viewed as processive. However, recent in vitro experiments with purified non-muscle myosin 2 (NM2) demonstrated myosin 2 filaments could move processively. Here, we establish processivity as a cellular property of NM2. Processive runs in central nervous system-derived CAD cells are most apparent as processive movements on bundled actin in protrusions that terminate at the leading edge. We find that processive velocities in vivo are consistent with in vitro measurements. NM2 makes these processive runs in its filamentous form against lamellipodia retrograde flow, though anterograde movement can still occur in the absence of actin dynamics. Comparing the processivity of NM2 isoforms, we find that NM2A moves slightly faster than NM2B. Finally, we demonstrate that this is not a cell-specific property, as we observe processive-like movements of NM2 in the lamella and subnuclear stress fibers of fibroblasts. Collectively, these observations further broaden NM2 functionality and the biological processes in which the already ubiquitous motor can contribute.
Collapse
Affiliation(s)
- Eric A Vitriol
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA
| | - Melissa A Quintanilla
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Joseph J Tidei
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Lee D Troughton
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Abigail Cody
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Bruno A Cisterna
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA
| | - Makenzie L Jane
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA
| | - Patrick W Oakes
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| | - Jordan R Beach
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL
| |
Collapse
|
17
|
Cholesterol esters form supercooled lipid droplets whose nucleation is facilitated by triacylglycerols. Nat Commun 2023; 14:915. [PMID: 36807572 PMCID: PMC9938224 DOI: 10.1038/s41467-023-36375-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 01/27/2023] [Indexed: 02/19/2023] Open
Abstract
Cellular cholesterol can be metabolized to its fatty acid esters, cholesteryl esters (CEs), to be stored in lipid droplets (LDs). With triacylglycerols (TGs), CEs represent the main neutral lipids in LDs. However, while TG melts at ~4 °C, CE melts at ~44 °C, raising the question of how CE-rich LDs form in cells. Here, we show that CE forms supercooled droplets when the CE concentration in LDs is above 20% to TG and, in particular, liquid-crystalline phases when the fraction of CEs is above 90% at 37 °C. In model bilayers, CEs condense and nucleate droplets when the CE/phospholipid ratio reaches over 10-15%. This concentration is reduced by TG pre-clusters in the membrane that thereby facilitate CE nucleation. Accordingly, blocking TG synthesis in cells is sufficient to strongly dampen CE LD nucleation. Finally, CE LDs emerged at seipins, which cluster and nucleate TG LDs in the ER. However, when TG synthesis is inhibited, similar numbers of LDs are generated in the presence and absence of seipin, suggesting that seipin controls CE LD formation via its TG clustering capacity. Our data point to a unique model whereby TG pre-clusters, favorable at seipins, catalyze the nucleation of CE LDs.
Collapse
|
18
|
Ismail VA, Naismith T, Kast DJ. The NTPase activity of the double FYVE domain-containing protein 1 regulates lipid droplet metabolism. J Biol Chem 2023; 299:102830. [PMID: 36574842 PMCID: PMC9881219 DOI: 10.1016/j.jbc.2022.102830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/26/2022] [Accepted: 12/08/2022] [Indexed: 12/25/2022] Open
Abstract
Lipid droplets (LDs) are transient lipid storage organelles that can be readily tapped to resupply cells with energy or lipid building blocks and therefore play a central role in cellular metabolism. However, the molecular factors and underlying mechanisms that regulate the growth and degradation of LDs are poorly understood. It has emerged that proteins that establish contacts between LDs and the endoplasmic reticulum play a critical role in regulating LD metabolism. Recently, the autophagy-related protein, double FYVE domain-containing protein 1 (DFCP1/ZFYVE1) was shown to reside at the interface of the endoplasmic reticulum and LDs, however, little is known about the involvement of DFCP1 in autophagy and LD metabolism. Here, we show that DFCP1 is a novel NTPase that regulates free fatty acid metabolism. Specifically, we show that DFPC1-knockdown, particularly during starvation, increases cellular free fatty acids and decreases the levels of cellular TAGs, resulting in accumulated small LDs. Using selective truncations, we demonstrate that DFCP1 accumulation on LDs in cells and in vitro is regulated by a previously unknown NTPase domain. Using spectroscopic approaches, we show that this NTPase domain can dimerize and can hydrolyze both ATP and GTP. Furthermore, mutations in DFCP1 that either impact nucleotide hydrolysis or dimerization result in changes in the accumulation of DFCP1 on LDs, changes in LD density and size, and colocalization of LDs to autophagosomes. Collectively, our findings suggest that DFCP1 is an NTPase that modulates the metabolism of LDs in cells.
Collapse
Affiliation(s)
- V A Ismail
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - T Naismith
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri, USA
| | - D J Kast
- Department of Cell Biology and Physiology, Washington University School of Medicine, St Louis, Missouri, USA.
| |
Collapse
|
19
|
Zhao P, Han H, Wu X, Wu J, Ren Z. ARP2/3 Regulates Fatty Acid Synthesis by Modulating Lipid Droplets' Motility. Int J Mol Sci 2022; 23:ijms23158730. [PMID: 35955862 PMCID: PMC9368945 DOI: 10.3390/ijms23158730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/04/2022] [Indexed: 11/16/2022] Open
Abstract
The breakdown of lipid droplets (LDs) provides energy and contributes to the proliferation and migration of cancer cells. Recent studies have suggested that motility plays a key role in LD breakdown. However, the molecular mechanisms underlying LD motility were poorly characterized. In this study, we examined the function of microfilament-associated proteins 2 and 3 (ARP2 and ARP3) in regulating LDs’ motility in Hela cells. ARP2/3 mediated the LDs’ physical contact with F-actin and promoted the recruitment of Myosin Heavy Chain 9 (MYH9). MYH9 regulated the LD content by binding with LDs and ARP2/3. The number of LDs and TG content was increased after MYH9 interfered. The genes related to FA-related genes and neutral lipid synthesis-related genes were significantly increased (p < 0.05) when ARP2 and ARP3 were overexpressed. Bioinformatic analysis indicated that the high expression of ARP2/3 was associated with a poorer prognosis in cervical squamous cell carcinoma (CSCC). This study showed the effect of cytoskeletal filaments on LD metabolism in cancer cells.
Collapse
Affiliation(s)
- Pengxiang Zhao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hongbo Han
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiang Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
- Hubei Hongshan Laboratory, Wuhan 430070, China
- Correspondence:
| |
Collapse
|
20
|
Pfisterer SG, Brock I, Kanerva K, Hlushchenko I, Paavolainen L, Ripatti P, Islam MM, Kyttälä A, Di Taranto MD, Scotto di Frega A, Fortunato G, Kuusisto J, Horvath P, Ripatti S, Laakso M, Ikonen E. Multiparametric platform for profiling lipid trafficking in human leukocytes. CELL REPORTS METHODS 2022; 2:100166. [PMID: 35474963 PMCID: PMC9017167 DOI: 10.1016/j.crmeth.2022.100166] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/26/2021] [Accepted: 01/18/2022] [Indexed: 11/25/2022]
Abstract
Systematic insight into cellular dysfunction can improve understanding of disease etiology, risk assessment, and patient stratification. We present a multiparametric high-content imaging platform enabling quantification of low-density lipoprotein (LDL) uptake and lipid storage in cytoplasmic droplets of primary leukocyte subpopulations. We validate this platform with samples from 65 individuals with variable blood LDL-cholesterol (LDL-c) levels, including familial hypercholesterolemia (FH) and non-FH subjects. We integrate lipid storage data into another readout parameter, lipid mobilization, measuring the efficiency with which cells deplete lipid reservoirs. Lipid mobilization correlates positively with LDL uptake and negatively with hypercholesterolemia and age, improving differentiation of individuals with normal and elevated LDL-c. Moreover, combination of cell-based readouts with a polygenic risk score for LDL-c explains hypercholesterolemia better than the genetic risk score alone. This platform provides functional insights into cellular lipid trafficking and has broad possible applications in dissecting the cellular basis of metabolic disorders.
Collapse
Affiliation(s)
- Simon G. Pfisterer
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Ivonne Brock
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Kristiina Kanerva
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Iryna Hlushchenko
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Lassi Paavolainen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Pietari Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Mohammad Majharul Islam
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
| | - Aija Kyttälä
- Finnish Institute for Health and Welfare (THL), THL Biobank, Helsinki, Finland
| | - Maria D. Di Taranto
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate scarl Naples, Napoli, Italy
| | | | - Giuliana Fortunato
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Napoli, Italy
- CEINGE Biotecnologie Avanzate scarl Naples, Napoli, Italy
| | - Johanna Kuusisto
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Peter Horvath
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Biological Research Center, Szeged, Hungary
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, Clinicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Markku Laakso
- Department of Medicine, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland
| | - Elina Ikonen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Haartmaninkatu 8, 00290 Helsinki, Finland
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
21
|
Liao PC, Yang EJ, Borgman T, Boldogh IR, Sing CN, Swayne TC, Pon LA. Touch and Go: Membrane Contact Sites Between Lipid Droplets and Other Organelles. Front Cell Dev Biol 2022; 10:852021. [PMID: 35281095 PMCID: PMC8908909 DOI: 10.3389/fcell.2022.852021] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/03/2022] [Indexed: 12/26/2022] Open
Abstract
Lipid droplets (LDs) have emerged not just as storage sites for lipids but as central regulators of metabolism and organelle quality control. These critical functions are achieved, in part, at membrane contact sites (MCS) between LDs and other organelles. MCS are sites of transfer of cellular constituents to or from LDs for energy mobilization in response to nutrient limitations, as well as LD biogenesis, expansion and autophagy. Here, we describe recent findings on the mechanisms underlying the formation and function of MCS between LDs and mitochondria, ER and lysosomes/vacuoles and the role of the cytoskeleton in promoting LD MCS through its function in LD movement and distribution in response to environmental cues.
Collapse
Affiliation(s)
- Pin-Chao Liao
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Emily J. Yang
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Taylor Borgman
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States
| | - Istvan R. Boldogh
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Cierra N. Sing
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States
| | - Theresa C. Swayne
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, United States
| | - Liza A. Pon
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, United States
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, United States
- *Correspondence: Liza A. Pon,
| |
Collapse
|
22
|
Zhao P, Jin Y, Wu X, Huang J, Chen L, Tan Y, Yuan H, Wu J, Ren Z. Artificial Lipid Droplets: Novel Effective Biomaterials to Protect Cells against Oxidative Stress and Lipotoxicity. NANOMATERIALS 2022; 12:nano12040672. [PMID: 35215001 PMCID: PMC8879118 DOI: 10.3390/nano12040672] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 02/01/2023]
Abstract
Lipid droplets (LDs) play an important role in the regulation of cellular stress. This suggests LDs can be applied as safe and effective biomaterials to alleviate cellular stress and lipotoxicity. Here, we constructed a convenient method to generate stable and pure artificial lipid droplets (aLDs). aLDs can maintain their biological function by incubating LD-associated proteins or organelles in vitro. It was validated that perilipin-coated aLDs could be uptaken by cells, significantly reducing hydrogen peroxide-induced reactive oxidative species (ROS) and alleviating cellular lipotoxicity caused by excess fatty acid. Our work demonstrated a direct role of LDs in regulating cellular stress levels, providing methods and potential value for future research and medical applications of LDs.
Collapse
Affiliation(s)
- Pengxiang Zhao
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
| | - Yi Jin
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
| | - Xiang Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
| | - Jin Huang
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
| | - Lupeng Chen
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
| | - Yanjie Tan
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
| | - Hong Yuan
- Key Laboratory of Pesticide and Chemical Biology of Ministry of Education, College of Chemistry, Central China Normal University, Wuhan 430079, China;
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan 430070, China; (P.Z.); (Y.J.); (X.W.); (J.H.); (L.C.); (Y.T.); (J.W.)
- Hubei Hongshan Laboratory, Wuhan 430072, China
- Correspondence:
| |
Collapse
|
23
|
Lv Y, Hong D, Fu L, Qian Y. NMMHC IIA triggered lipid metabolize reprogramming resulting in vascular endothelial cellular tight junction injury. Mol Biol Rep 2022; 49:2805-2819. [PMID: 35064404 DOI: 10.1007/s11033-021-07092-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/16/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND PURPOSE Nonmuscle myosin heavy chain IIA, played an essential role in the promotion of tight junction injury in vascular endothelial cells under oxygen glucose deprivation condition. Rat microvascular endothelial cells had been confirmed to have the susceptibility to ox-LDL stimulation under OGD condition. We proposed the hypothesis that lipid metabolic reprogramming might be the root cause for damage to RBMCs tight junction. METHODS Untargeted shotgun and targeted lipid metabolomics mass spectrometry approaches combined with principal component analysis was applied to better define the lipids contributing to the variance observed between control and different OGD time. The protein expression of tight junction of RBMCs: occludin, claudin-5, and ZO-1 were detected with immunofluorescence staining and western blot. The proof of the interaction between NMMHC IIA and SREBP1 was investigated via GST-pull down, while their specific binding fragments were also confirmed. The regulation mechanism of NMMHC IIA on SREBP1 was investigated to explore downstream regulatory signaling pathways. RESULTS Untargeted and targeted shotgun lipidomics data revealed that OGD might be the conditional factor in reshaping lipid components. Mechanistic studies showed that with the increase of OGD time, PCA analysis of lipidomics obtained from RBMCs indicated their specificity in reshaping lipid components, while ≥80% major lipid components phospholipids and sphingolipids transferred from phospholipids, sphingolipids, and neutral lipids, of which neutral lipids taken the largest proportion with OGD time course. Perturbing reprogramming of lipid composition was less susceptible to OGD condition via knockdown of NMMHC IIA of vascular endothelial cells. Knockdown of NMMHC IIA could promote tight junction defense to OGD condition. NMMHC IIA could directly bind with SREBP1, then could affect sterol regulatory element binding protein-1 to adjust lipid metabolize reprogramming of RBMCs. CONCLUSIONS Mechanistic studies showed that perturbing reprogramming of lipid composition could enhance tight junction damage, which was mediated by the opposing effects of NMMHC IIA.
Collapse
Affiliation(s)
- Yanni Lv
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Jiangxi, 330006, China.
| | - Daojun Hong
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Jiangxi, 330006, China
| | - Longsheng Fu
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Jiangxi, 330006, China
| | - Yisong Qian
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Jiangxi, 330006, China
- Institute of Translational Medicine, Nanchang University, Nanchang, 330001, China
| |
Collapse
|
24
|
Tan Y, Jin Y, Zhao P, Wu J, Ren Z. Lipid droplets contribute myogenic differentiation in C2C12 by promoting the remodeling of the acstin-filament. Cell Death Dis 2021; 12:1102. [PMID: 34815388 PMCID: PMC8611090 DOI: 10.1038/s41419-021-04273-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/17/2021] [Accepted: 10/04/2021] [Indexed: 12/29/2022]
Abstract
Lipid droplet (LD), a multi-functional organelle, is found in most eukaryotic cells. LDs participate in the regulation of many cellular processes including proliferation, stress, and apoptosis. Previous studies showed the athlete's paradox that trained athletes accumulate LDs in their skeletal muscle. However, the impact of LDs on skeletal muscle and myogenesis is not clear. We discovered that C2C12 myoblast cells containing more LDs formed more multinucleated muscle fibers. We also discovered that LDs promoted cell migration and fusion by promoting actin-filaments remodeling. Mechanistically, two LD-proteins, Acyl-CoA synthetase long chain family member 3 (ACSL3) and lysophosphatidylcholine acyltransferase 1 (LPCAT1), medicated the recruitment of actinin proteins which contributed to actin-filaments formation on the surface of LDs. During remodeling, the actinin proteins on LDs surface translocated to actin-filaments via ARF1/COPI vesicles. Our study demonstrate LDs contribute to cell differentiation, which lead to new insight into the LD function.
Collapse
Affiliation(s)
- Yanjie Tan
- grid.35155.370000 0004 1790 4137Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, 430070 Wuhan, Hubei P. R. China ,grid.410585.d0000 0001 0495 1805Institute of Biomedical Sciences, Key Laboratory of Animal Resistance Biology of Shandong Province, College of Life Sciences, Shandong Normal University, 250014 Jinan, Shandong China
| | - Yi Jin
- grid.35155.370000 0004 1790 4137Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, 430070 Wuhan, Hubei P. R. China
| | - Pengxiang Zhao
- grid.35155.370000 0004 1790 4137Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, 430070 Wuhan, Hubei P. R. China
| | - Jian Wu
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, 430070, Wuhan, Hubei, P. R. China.
| | - Zhuqing Ren
- Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of the Ministry of Education & Key Laboratory of Swine Genetics and Breeding of the Ministry of Agriculture and Rural Affairs, College of Animal Science, Huazhong Agricultural University, 430070, Wuhan, Hubei, P. R. China. .,Hubei Hongshan Laboratory, Wuhan, Hubei, China.
| |
Collapse
|
25
|
Takahashi K, Kanerva K, Vanharanta L, Almeida‐Souza L, Lietha D, Olkkonen VM, Ikonen E. ORP2 couples LDL-cholesterol transport to FAK activation by endosomal cholesterol/PI(4,5)P 2 exchange. EMBO J 2021; 40:e106871. [PMID: 34124795 PMCID: PMC8281050 DOI: 10.15252/embj.2020106871] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 12/19/2022] Open
Abstract
Low-density lipoprotein (LDL)-cholesterol delivery from late endosomes to the plasma membrane regulates focal adhesion dynamics and cell migration, but the mechanisms controlling it are poorly characterized. Here, we employed auxin-inducible rapid degradation of oxysterol-binding protein-related protein 2 (ORP2/OSBPL2) to show that endogenous ORP2 mediates the transfer of LDL-derived cholesterol from late endosomes to focal adhesion kinase (FAK)-/integrin-positive recycling endosomes in human cells. In vitro, cholesterol enhances membrane association of FAK to PI(4,5)P2 -containing lipid bilayers. In cells, ORP2 stimulates FAK activation and PI(4,5)P2 generation in endomembranes, enhancing cell adhesion. Moreover, ORP2 increases PI(4,5)P2 in NPC1-containing late endosomes in a FAK-dependent manner, controlling their tubulovesicular trafficking. Together, these results provide evidence that ORP2 controls FAK activation and LDL-cholesterol plasma membrane delivery by promoting bidirectional cholesterol/PI(4,5)P2 exchange between late and recycling endosomes.
Collapse
Affiliation(s)
- Kohta Takahashi
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
- Present address:
Laboratory of Microbiology and ImmunologyGraduate School of Pharmaceutical SciencesChiba UniversityChibaJapan
| | - Kristiina Kanerva
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Lauri Vanharanta
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Leonardo Almeida‐Souza
- Helsinki Institute of Life Science, HiLIFEUniversity of HelsinkiHelsinkiFinland
- Faculty of Biological and Environmental SciencesUniversity of HelsinkiHelsinkiFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| | - Daniel Lietha
- Centro de Investigaciones Biológicas Margarita Salas (CIB)Spanish National Research Council (CSIC)MadridSpain
| | - Vesa M Olkkonen
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research ProgramFaculty of MedicineUniversity of HelsinkiHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| |
Collapse
|
26
|
Monson EA, Whelan DR, Helbig KJ. Lipid Droplet Motility Increases Following Viral Immune Stimulation. Int J Mol Sci 2021; 22:4418. [PMID: 33922664 PMCID: PMC8122965 DOI: 10.3390/ijms22094418] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/20/2021] [Accepted: 04/21/2021] [Indexed: 12/15/2022] Open
Abstract
Lipid droplets (LDs) have traditionally been thought of as solely lipid storage compartments for cells; however, in the last decade, they have emerged as critical organelles in health and disease. LDs are highly dynamic within cells, and their movement is critical in organelle-organelle interactions. Their dynamics are known to change during cellular stress or nutrient deprivation; however, their movement during pathogen infections, especially at very early timepoints, is under-researched. This study aimed to track LD dynamics in vitro, in an astrocytic model of infection. Cells were either stimulated with a dsRNA viral mimic, poly I:C, or infected with the RNA virus, Zika virus. Individual LDs within infected cells were analysed to determine displacement and speed, and average LD characteristics for multiple individual cells calculated. Both LD displacement and mean speed were significantly enhanced in stimulated cells over a time course of infection with an increase seen as early as 2 h post-infection. With the emerging role for LDs during innate host responses, understanding their dynamics is critical to elucidate how these organelles influence the outcome of viral infection.
Collapse
Affiliation(s)
- Ebony A. Monson
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne 3086, Australia;
| | - Donna R. Whelan
- Department of Pharmacy and Biomedical Sciences, La Trobe Institute for Molecular Science, La Trobe University, Melbourne 3086, Australia
| | - Karla J. Helbig
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University, Melbourne 3086, Australia;
| |
Collapse
|
27
|
Motility Plays an Important Role in the Lifetime of Mammalian Lipid Droplets. Int J Mol Sci 2021; 22:ijms22083802. [PMID: 33916886 PMCID: PMC8067576 DOI: 10.3390/ijms22083802] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/25/2021] [Accepted: 04/01/2021] [Indexed: 01/31/2023] Open
Abstract
The lipid droplet is a kind of organelle that stores neutral lipids in cells. Recent studies have found that in addition to energy storage, lipid droplets also play an important role in biological processes such as resistance to stress, immunity, cell proliferation, apoptosis, and signal transduction. Lipid droplets are formed at the endoplasmic reticulum, and mature lipid droplets participate in various cellular processes. Lipid droplets are decomposed by lipase and lysosomes. In the life of a lipid droplet, the most important thing is to interact with other organelles, including the endoplasmic reticulum, mitochondria, peroxisomes, and autophagic lysosomes. The interaction between lipid droplets and other organelles requires them to be close to each other, which inevitably involves the motility of lipid droplets. In fact, through many microscopic observation techniques, researchers have discovered that lipid droplets are highly dynamic organelles that move quickly. This paper reviews the process of lipid droplet motility, focusing on explaining the molecular basis of lipid droplet motility, the factors that regulate lipid droplet motility, and the influence of motility on the formation and decomposition of lipid droplets. In addition, this paper also proposes several unresolved problems for lipid droplet motility. Finally, this paper makes predictions about the future research of lipid droplet motility.
Collapse
|
28
|
Alajbegovic A, Holmberg J, Daoud F, Rippe C, Kalliokoski G, Ekman M, Daudi S, Ragnarsson S, Swärd K, Albinsson S. MRTFA overexpression promotes conversion of human coronary artery smooth muscle cells into lipid-laden foam cells. Vascul Pharmacol 2021; 138:106837. [PMID: 33516965 DOI: 10.1016/j.vph.2021.106837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/22/2020] [Accepted: 01/21/2021] [Indexed: 01/26/2023]
Abstract
OBJECTIVE Smooth muscle cells contribute significantly to lipid-laden foam cells in atherosclerotic plaques. However, the underlying mechanisms transforming smooth muscle cells into foam cells are poorly understood. The purpose of this study was to gain insight into the molecular mechanisms regulating smooth muscle foam cell formation. APPROACH AND RESULTS Using human coronary artery smooth muscle cells we found that the transcriptional co-activator MRTFA promotes lipid accumulation via several mechanisms, including direct transcriptional control of LDL receptor, enhanced fluid-phase pinocytosis and reduced lipid efflux. Inhibition of MRTF activity with CCG1423 and CCG203971 significantly reduced lipid accumulation. Furthermore, we demonstrate enhanced MRTFA expression in vascular remodeling of human vessels. CONCLUSIONS This study demonstrates a novel role for MRTFA as an important regulator of lipid homeostasis in vascular smooth muscle cells. Thus, MRTFA could potentially be a new therapeutic target for inhibition of vascular lipid accumulation.
Collapse
Affiliation(s)
- Azra Alajbegovic
- Department of Experimental Medical Science, Lund University, Sweden.
| | - Johan Holmberg
- Department of Experimental Medical Science, Lund University, Sweden
| | - Fatima Daoud
- Department of Experimental Medical Science, Lund University, Sweden
| | - Catarina Rippe
- Department of Experimental Medical Science, Lund University, Sweden
| | | | - Mari Ekman
- Department of Experimental Medical Science, Lund University, Sweden
| | - Sébastien Daudi
- Department of Clinical Science, Lund University, Lund, Sweden
| | | | - Karl Swärd
- Department of Experimental Medical Science, Lund University, Sweden
| | | |
Collapse
|
29
|
Prasanna X, Salo VT, Li S, Ven K, Vihinen H, Jokitalo E, Vattulainen I, Ikonen E. Seipin traps triacylglycerols to facilitate their nanoscale clustering in the endoplasmic reticulum membrane. PLoS Biol 2021; 19:e3000998. [PMID: 33481779 PMCID: PMC7857593 DOI: 10.1371/journal.pbio.3000998] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 02/03/2021] [Accepted: 11/02/2020] [Indexed: 11/19/2022] Open
Abstract
Seipin is a disk-like oligomeric endoplasmic reticulum (ER) protein important for lipid droplet (LD) biogenesis and triacylglycerol (TAG) delivery to growing LDs. Here we show through biomolecular simulations bridged to experiments that seipin can trap TAGs in the ER bilayer via the luminal hydrophobic helices of the protomers delineating the inner opening of the seipin disk. This promotes the nanoscale sequestration of TAGs at a concentration that by itself is insufficient to induce TAG clustering in a lipid membrane. We identify Ser166 in the α3 helix as a favored TAG occupancy site and show that mutating it compromises the ability of seipin complexes to sequester TAG in silico and to promote TAG transfer to LDs in cells. While the S166D-seipin mutant colocalizes poorly with promethin, the association of nascent wild-type seipin complexes with promethin is promoted by TAGs. Together, these results suggest that seipin traps TAGs via its luminal hydrophobic helices, serving as a catalyst for seeding the TAG cluster from dissolved monomers inside the seipin ring, thereby generating a favorable promethin binding interface.
Collapse
Affiliation(s)
- Xavier Prasanna
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Veijo T. Salo
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Shiqian Li
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Katharina Ven
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Helena Vihinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Eija Jokitalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Elina Ikonen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
- * E-mail:
| |
Collapse
|
30
|
Zhang MF, Li QL, Yang YF, Cao Y, Zhang CZ. FMNL1 Exhibits Pro-Metastatic Activity via CXCR2 in Clear Cell Renal Cell Carcinoma. Front Oncol 2020; 10:564614. [PMID: 33324547 PMCID: PMC7726248 DOI: 10.3389/fonc.2020.564614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 11/02/2020] [Indexed: 12/25/2022] Open
Abstract
Formin-like (FMNL) proteins are responsible for cytoskeletal remodeling and have been implicated in the progression and spread of human cancers. Yet the clinical significance and biological function of FMNL1 in clear cell renal cell carcinoma (ccRCC) remain unclear. In this study, the expression of FMNL1 in ccRCC and its clinical value were determined by tissue microarray-based IHC and statistical analyses. The role of FMNL1 in ccRCC metastasis and the underlying mechanism were investigated via in vitro and in vivo models using gene regulation detection, ChIP, Luciferase reporter assays, and rescue experiments. We show that FMNL1 is upregulated in ccRCC and exhibits pro-metastatic activity via induction of CXCR2. High expression of FMNL1 is significantly correlated with advanced tumor stage, higher pathological tumor grade, tumor metastasis, and unfavorable prognosis in two independent cohorts containing over 800 patients with ccRCC. The upregulation of FMNL1 in ccRCC is mediated by the loss of GATA3. Ectopic expression of FMNL1 promotes, whereas FMNL1 depletion inhibits cell migration in vitro and tumor metastasis in vivo. The FMNL1-enhanced cell mobility is markedly attenuated by the knockdown of CXCR2. Further studies demonstrate that FMNL1 increases the expression of CXCR2 via HDAC1. In clinical samples, FMNL1 expression is positively associated with CXCR2, and is negatively connected to GATA3 expression. Collectively, our data suggest FMNL1 serve as a potential prognostic factor and function as an oncogene. The axis of GATA3/FMNL1/CXCR2 may present a promising therapeutic target for tumor metastasis in ccRCC.
Collapse
Affiliation(s)
- Mei-Fang Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiu-Li Li
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yu-Feng Yang
- Department of Pathology, Dongguan Third People's Hospital, Dongguan, China
| | - Yun Cao
- Department of Pathology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chris Zhiyi Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
31
|
Friend or Foe: Lipid Droplets as Organelles for Protein and Lipid Storage in Cellular Stress Response, Aging and Disease. Molecules 2020; 25:molecules25215053. [PMID: 33143278 PMCID: PMC7663626 DOI: 10.3390/molecules25215053] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 02/06/2023] Open
Abstract
Lipid droplets (LDs) were considered as a mere lipid storage organelle for a long time. Recent evidence suggests that LDs are in fact distinct and dynamic organelles with a specialized proteome and functions in many cellular roles. As such, LDs contribute to cellular signaling, protein and lipid homeostasis, metabolic diseases and inflammation. In line with the multitude of functions, LDs interact with many cellular organelles including mitochondria, peroxisomes, lysosomes, the endoplasmic reticulum and the nucleus. LDs are highly mobile and dynamic organelles and impaired motility disrupts the interaction with other organelles. The reduction of interorganelle contacts results in a multitude of pathophysiologies and frequently in neurodegenerative diseases. Contacts not only supply lipids for β-oxidation in mitochondria and peroxisomes, but also may include the transfer of toxic lipids as well as misfolded and harmful proteins to LDs. Furthermore, LDs assist in the removal of protein aggregates when severe proteotoxic stress overwhelms the proteasomal system. During imbalance of cellular lipid homeostasis, LDs also support cellular detoxification. Fine-tuning of LD function is of crucial importance and many diseases are associated with dysfunctional LDs. We summarize the current understanding of LDs and their interactions with organelles, providing a storage site for harmful proteins and lipids during cellular stress, aging inflammation and various disease states.
Collapse
|
32
|
Zhang M, Han Y, Zhai Y, Ma X, An X, Zhang S, Li Z. Integrative analysis of circRNAs, miRNAs, and mRNAs profiles to reveal ceRNAs networks in chicken intramuscular and abdominal adipogenesis. BMC Genomics 2020; 21:594. [PMID: 32847498 PMCID: PMC7450580 DOI: 10.1186/s12864-020-07000-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tissue-specific fat deposition is regulated by a series of complex regulatory mechanisms. Reports indicate that epigenetic regulators, such as circular RNAs (circRNAs), are crucial in diseases progression, animal development, metabolism, and adipogenesis. In this study, to assess the functional roles of circRNAs in adipogenesis and tissue-specific fat deposition, we comprehensively analyzed the Ribo-Zero RNA-Seq and miRNAs data during chicken intramuscular and abdominal adipogenic differentiation. RESULTS circRNAs and miRNAs profiles during chicken adipogenic differentiation were found in adipocytes derived from various adipose tissues. It was also discovered that high levels of downregulated miRNAs potentially promote adipogenesis by activating their target genes which are associated with fatty acid metabolism and adipogenic differentiation. Through analysis of the correlation between the expression levels of circRNAs and adipogenic genes, as well as the dynamic expression patterns of circRNAs during adipogenic differentiation, several candidate circRNAs were identified. Moreover, competing endogenous RNA (ceRNAs) networks were constructed during chicken intramuscular and abdominal adipogenesis by combining miRNAs with mRNAs data. Several candidate circRNAs potentially influence adipogenesis by regulating miRNAs via PPAR and fatty acid metabolism-related pathways were identified, such as circLCLAT1, circFNDC3AL, circCLEC19A and circARMH1. CONCLUSION In conclusion, our findings reveal that circRNAs and the circRNA-miRNAs-mRNAs-ceRNAs network may play important roles in chicken adipocytes differentiation and tissue-specific fat deposition.
Collapse
Affiliation(s)
- Meng Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, 130021, Jilin, China
| | - Yu Han
- College of Veterinary Medicine, Jilin University, Changchun, 130021, Jilin, China
| | - Yanhui Zhai
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, 130021, Jilin, China
| | - Xiangfei Ma
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450002, Henan, China
| | - Xinglan An
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, 130021, Jilin, China
| | - Sheng Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, 130021, Jilin, China.
| | - Ziyi Li
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital, Jilin University, Changchun, 130021, Jilin, China.
| |
Collapse
|
33
|
Vanharanta L, Peränen J, Pfisterer SG, Enkavi G, Vattulainen I, Ikonen E. High‐content imaging and structure‐based predictions reveal functional differences between Niemann‐Pick C1 variants. Traffic 2020; 21:386-397. [DOI: 10.1111/tra.12727] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Lauri Vanharanta
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of MedicineUniversity of Helsinki Helsinki Finland
- Minerva Foundation Institute for Medical Research Helsinki Finland
| | - Johan Peränen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of MedicineUniversity of Helsinki Helsinki Finland
- Minerva Foundation Institute for Medical Research Helsinki Finland
| | - Simon G. Pfisterer
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of MedicineUniversity of Helsinki Helsinki Finland
| | - Giray Enkavi
- Department of PhysicsUniversity of Helsinki Helsinki Finland
- Computational Physics LaboratoryTampere University of Technology Tampere Finland
| | - Ilpo Vattulainen
- Department of PhysicsUniversity of Helsinki Helsinki Finland
- Computational Physics LaboratoryTampere University of Technology Tampere Finland
| | - Elina Ikonen
- Department of Anatomy and Stem Cells and Metabolism Research Program, Faculty of MedicineUniversity of Helsinki Helsinki Finland
- Minerva Foundation Institute for Medical Research Helsinki Finland
| |
Collapse
|
34
|
Chen F, Yin Y, Chua BT, Li P. CIDE family proteins control lipid homeostasis and the development of metabolic diseases. Traffic 2019; 21:94-105. [PMID: 31746121 DOI: 10.1111/tra.12717] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/03/2019] [Accepted: 11/15/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Feng‐Jung Chen
- Institute of Metabolism and Integrative Biology, the Human Phenome InstituteFudan University, and Zhongshan Hospital of Fudan University Shanghai China
| | - Yesheng Yin
- Institute of Metabolism and Integrative Biology, the Human Phenome InstituteFudan University, and Zhongshan Hospital of Fudan University Shanghai China
| | - Boon Tin Chua
- Institute of Metabolism and Integrative Biology, the Human Phenome InstituteFudan University, and Zhongshan Hospital of Fudan University Shanghai China
| | - Peng Li
- State Key Laboratory of Membrane Biology and Tsinghua‐Peking Center for Life Sciences, Beijing Advanced Innovation Center for Structural Biology, School of Life SciencesTsinghua University Beijing China
| |
Collapse
|
35
|
Abstract
Lipid droplets (LDs) are fat storage organelles integral to energy homeostasis and a wide range of cellular processes. LDs physically and functionally interact with many partner organelles, including the ER, mitochondria, lysosomes, and peroxisomes. Recent findings suggest that the dynamics of LD inter-organelle contacts is in part controlled by LD intracellular motility. LDs can be transported directly by motor proteins along either actin filaments or microtubules, via Kinesin-1, Cytoplasmic Dynein, and type V Myosins. LDs can also be propelled indirectly, by hitchhiking on other organelles, cytoplasmic flows, and potentially actin polymerization. Although the anchors that attach motors to LDs remain elusive, other regulators of LD motility have been identified, ranging from modification of the tracks to motor co-factors to members of the perilipin family of LD proteins. Manipulating these regulatory pathways provides a tool to probe whether altered motility affects organelle contacts and has revealed that LD motility can promote interactions with numerous partners, with profound consequences for metabolism. LD motility can cause dramatic redistribution of LDs between a clustered and a dispersed state, resulting in altered organelle contacts and LD turnover. We propose that LD motility can thus promote switches in the metabolic state of a cell. Finally, LD motility is also important for LD allocation during cell division. In a number of animal embryos, uneven allocation results in a large difference in LD content in distinct daughter cells, suggesting cell-type specific LD needs.
Collapse
Affiliation(s)
- Marcus D Kilwein
- Department of Biology, University of Rochester, RC Box 270211, Rochester, NY 14627, USA
| | - M A Welte
- Department of Biology, University of Rochester, RC Box 270211, Rochester, NY 14627, USA
| |
Collapse
|
36
|
Abstract
Myosin 2 plays a central role in numerous, fundamental, actin-based biological processes, including cell migration, cell division, and the adhesion of cells to substrates and other cells. Here, we highlight recent studies in which the forces created by actomyosin 2 have been shown to also impact tension-sensitive ion channels and cell metabolism.
Collapse
Affiliation(s)
- Melissa A Quintanilla
- Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Center for Translational Research and Education, Maywood, IL, USA
| | - John A Hammer
- Cell and Developmental Biology Center, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jordan R Beach
- Cell and Molecular Physiology, Loyola University Chicago, Stritch School of Medicine, Center for Translational Research and Education, Maywood, IL, USA
| |
Collapse
|
37
|
Heybrock S, Kanerva K, Meng Y, Ing C, Liang A, Xiong ZJ, Weng X, Ah Kim Y, Collins R, Trimble W, Pomès R, Privé GG, Annaert W, Schwake M, Heeren J, Lüllmann-Rauch R, Grinstein S, Ikonen E, Saftig P, Neculai D. Lysosomal integral membrane protein-2 (LIMP-2/SCARB2) is involved in lysosomal cholesterol export. Nat Commun 2019; 10:3521. [PMID: 31387993 PMCID: PMC6684646 DOI: 10.1038/s41467-019-11425-0] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 07/12/2019] [Indexed: 11/30/2022] Open
Abstract
The intracellular transport of cholesterol is subject to tight regulation. The structure of the lysosomal integral membrane protein type 2 (LIMP-2, also known as SCARB2) reveals a large cavity that traverses the molecule and resembles the cavity in SR-B1 that mediates lipid transfer. The detection of cholesterol within the LIMP-2 structure and the formation of cholesterol-like inclusions in LIMP-2 knockout mice suggested the possibility that LIMP2 transports cholesterol in lysosomes. We present results of molecular modeling, crosslinking studies, microscale thermophoresis and cell-based assays that support a role of LIMP-2 in cholesterol transport. We show that the cavity in the luminal domain of LIMP-2 can bind and deliver exogenous cholesterol to the lysosomal membrane and later to lipid droplets. Depletion of LIMP-2 alters SREBP-2-mediated cholesterol regulation, as well as LDL-receptor levels. Our data indicate that LIMP-2 operates in parallel with Niemann Pick (NPC)-proteins, mediating a slower mode of lysosomal cholesterol export.
Collapse
Affiliation(s)
- Saskia Heybrock
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany
| | - Kristiina Kanerva
- Faculty of Medicine, Anatomy and Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Ying Meng
- Department of Cell Biology, and Department of Pathology Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Chris Ing
- Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Anna Liang
- Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Zi-Jian Xiong
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Xialian Weng
- Department of Cell Biology, and Department of Pathology Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Young Ah Kim
- Department of Chemistry and Biochemistry, Queens College, City University of New York, Flushing, New York, USA
| | - Richard Collins
- Cell Biology Program, Hospital for Sick Children, Toronto, M5G 1X8, Canada
| | - William Trimble
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
- Cell Biology Program, Hospital for Sick Children, Toronto, M5G 1X8, Canada
- Department of Physiology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Régis Pomès
- Program in Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
| | - Gilbert G Privé
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada
- Princes Margaret Cancer Centre, Toronto, ON, Canada
| | - Wim Annaert
- Laboratory for Membrane Trafficking, VIB-Center for Brain and Disease Research, Leuven, Belgium
| | - Michael Schwake
- Faculty of Chemistry, Biochemistry III, University of Bielefeld, 33615, Bielefeld, Germany
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Joerg Heeren
- Institut für Biochemie und Molekulare Zellbiologie, Zentrum für Experimentelle Medizin, Universitätsklinikum Hamburg-Eppendorf, Hamburg-Eppendorf, Germany
| | | | - Sergio Grinstein
- Department of Biochemistry, University of Toronto, Toronto, M5S 1A8, Canada.
- Cell Biology Program, Hospital for Sick Children, Toronto, M5G 1X8, Canada.
| | - Elina Ikonen
- Faculty of Medicine, Anatomy and Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland.
- Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| | - Paul Saftig
- Biochemisches Institut, Christian-Albrechts-Universität Kiel, Kiel, Germany.
| | - Dante Neculai
- Department of Cell Biology, and Department of Pathology Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
38
|
Salo VT, Li S, Vihinen H, Hölttä-Vuori M, Szkalisity A, Horvath P, Belevich I, Peränen J, Thiele C, Somerharju P, Zhao H, Santinho A, Thiam AR, Jokitalo E, Ikonen E. Seipin Facilitates Triglyceride Flow to Lipid Droplet and Counteracts Droplet Ripening via Endoplasmic Reticulum Contact. Dev Cell 2019; 50:478-493.e9. [PMID: 31178403 DOI: 10.1016/j.devcel.2019.05.016] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 02/27/2019] [Accepted: 05/03/2019] [Indexed: 01/02/2023]
Abstract
Seipin is an oligomeric integral endoplasmic reticulum (ER) protein involved in lipid droplet (LD) biogenesis. To study the role of seipin in LD formation, we relocalized it to the nuclear envelope and found that LDs formed at these new seipin-defined sites. The sites were characterized by uniform seipin-mediated ER-LD necks. At low seipin content, LDs only grew at seipin sites, and tiny, growth-incompetent LDs appeared in a Rab18-dependent manner. When seipin was removed from ER-LD contacts within 1 h, no lipid metabolic defects were observed, but LDs became heterogeneous in size. Studies in seipin-ablated cells and model membranes revealed that this heterogeneity arises via a biophysical ripening process, with triglycerides partitioning from smaller to larger LDs through droplet-bilayer contacts. These results suggest that seipin supports the formation of structurally uniform ER-LD contacts and facilitates the delivery of triglycerides from ER to LDs. This counteracts ripening-induced shrinkage of small LDs.
Collapse
Affiliation(s)
- Veijo T Salo
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Shiqian Li
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Helena Vihinen
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maarit Hölttä-Vuori
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | | | | - Ilya Belevich
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Johan Peränen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | | | - Pentti Somerharju
- Department of Biochemistry, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Hongxia Zhao
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Alexandre Santinho
- Laboratoire de Physique de l'Ecole Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Universite de Paris, Paris, France
| | - Abdou Rachid Thiam
- Laboratoire de Physique de l'Ecole Normale Supérieure, ENS, Université PSL, CNRS, Sorbonne Université, Universite de Paris, Paris, France.
| | - Eija Jokitalo
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland.
| | - Elina Ikonen
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland.
| |
Collapse
|
39
|
Menon D, Singh K, Pinto SM, Nandy A, Jaisinghani N, Kutum R, Dash D, Prasad TSK, Gandotra S. Quantitative Lipid Droplet Proteomics Reveals Mycobacterium tuberculosis Induced Alterations in Macrophage Response to Infection. ACS Infect Dis 2019; 5:559-569. [PMID: 30663302 PMCID: PMC6466475 DOI: 10.1021/acsinfecdis.8b00301] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
![]()
Growing
evidence suggests the importance of lipid metabolism in pathogenesis
of tuberculosis. Neutral lipids form the majority of lipids in a caseous
granuloma, a pathology characteristic of tuberculosis. Cytosolic lipid
droplets (LDs) of macrophages form the store house of these lipids
and have been demonstrated to contribute to the inflammatory response
to infection. The proteome of lipid droplets reflects the mechanisms
of lipid metabolism active under a condition. However, infection induced
changes in the proteome of these dynamic organelles remains elusive.
Here, we employed quantitative proteomics to identify alterations
induced upon infection with live Mycobacterium tuberculosis (Mtb) in comparison with heat killed bacilli or uninfected macrophages.
We found increased abundance of proteins coupled with lipid metabolism,
protein synthesis, and vesicular transport function in LDs upon infection
with live Mtb. Using biochemical methods and microscopy, we validated
ADP-ribosyltransferase (Arf)-like 8 (ARL8B) to be increased on the
lipid droplet surface of live Mtb infected macrophages and that ARL8B
is a bonafide LD protein. This study provides the first proteomic
evidence that the dynamic responses to infection also encompass changes
at the level of LDs. This information will be important in understanding
how Mtb manipulates lipid metabolism and defense mechanisms of the
host macrophage.
Collapse
Affiliation(s)
- Dilip Menon
- Cardiorespiratory Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kaurab Singh
- Cardiorespiratory Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sneha M. Pinto
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Ananya Nandy
- Cardiorespiratory Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Neetika Jaisinghani
- Cardiorespiratory Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rintu Kutum
- Informatics and Big Data, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Debasis Dash
- Informatics and Big Data, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Center, Yenepoya (Deemed to be University), Mangalore 575018, India
- Institute of Bioinformatics, International Technology Park, Bangalore 560066, India
| | - Sheetal Gandotra
- Cardiorespiratory Disease Biology, CSIR-Institute of Genomics and Integrative Biology, Sukhdev Vihar, Mathura Road, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
40
|
Xu H, Zhang H, Liu G, Kong L, Zhu X, Tian X, Zhang Z, Zhang R, Wu Z, Tian Y, Zhou H. Coumarin-Based Fluorescent Probes for Super-resolution and Dynamic Tracking of Lipid Droplets. Anal Chem 2018; 91:977-982. [DOI: 10.1021/acs.analchem.8b04079] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
41
|
Salo VT, Ikonen E. Moving out but keeping in touch: contacts between endoplasmic reticulum and lipid droplets. Curr Opin Cell Biol 2018; 57:64-70. [PMID: 30476754 DOI: 10.1016/j.ceb.2018.11.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/06/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022]
Abstract
The formation of neutral lipid filled and phospholipid monolayer engulfed lipid droplets (LDs) from the bilayer of the endoplasmic reticulum (ER) is an active area of investigation. This process harnesses the biophysical properties of the lipids involved and necessitates cooperation of protein machineries in both organelle membranes. Increasing evidence suggests that once formed, LDs keep close contact to the mother organelle and that this may be achieved via several, morphologically distinct and potentially functionally specialized connections. These may help LDs to dynamically respond to changes in lipid metabolic status sensed by the ER. In this review, we will discuss recent progress in understanding how LDs interact with the ER.
Collapse
Affiliation(s)
- Veijo T Salo
- Faculty of Medicine, Dept. of Anatomy and HiLIFE, Univ. of Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Elina Ikonen
- Faculty of Medicine, Dept. of Anatomy and HiLIFE, Univ. of Helsinki, Finland; Minerva Foundation Institute for Medical Research, Helsinki, Finland
| |
Collapse
|
42
|
Zhang C, Liu P. The New Face of the Lipid Droplet: Lipid Droplet Proteins. Proteomics 2018; 19:e1700223. [DOI: 10.1002/pmic.201700223] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/13/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Congyan Zhang
- National Laboratory of BiomacromoleculesCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of Sciences Beijing 100101 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Pingsheng Liu
- National Laboratory of BiomacromoleculesCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of Sciences Beijing 100101 China
- University of Chinese Academy of Sciences Beijing 100049 China
| |
Collapse
|
43
|
Tharp KM, Kang MS, Timblin GA, Dempersmier J, Dempsey GE, Zushin PJH, Benavides J, Choi C, Li CX, Jha AK, Kajimura S, Healy KE, Sul HS, Saijo K, Kumar S, Stahl A. Actomyosin-Mediated Tension Orchestrates Uncoupled Respiration in Adipose Tissues. Cell Metab 2018; 27:602-615.e4. [PMID: 29514068 PMCID: PMC5897043 DOI: 10.1016/j.cmet.2018.02.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 10/18/2017] [Accepted: 02/06/2018] [Indexed: 12/17/2022]
Abstract
The activation of brown/beige adipose tissue (BAT) metabolism and the induction of uncoupling protein 1 (UCP1) expression are essential for BAT-based strategies to improve metabolic homeostasis. Here, we demonstrate that BAT utilizes actomyosin machinery to generate tensional responses following adrenergic stimulation, similar to muscle tissues. The activation of actomyosin mechanics is critical for the acute induction of oxidative metabolism and uncoupled respiration in UCP1+ adipocytes. Moreover, we show that actomyosin-mediated elasticity regulates the thermogenic capacity of adipocytes via the mechanosensitive transcriptional co-activators YAP and TAZ, which are indispensable for normal BAT function. These biomechanical signaling mechanisms may inform future strategies to promote the expansion and activation of brown/beige adipocytes.
Collapse
Affiliation(s)
- Kevin M Tharp
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michael S Kang
- UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Greg A Timblin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jon Dempersmier
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Garret E Dempsey
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Peter-James H Zushin
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jaime Benavides
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Catherine Choi
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Catherine X Li
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Amit K Jha
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Shingo Kajimura
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kevin E Healy
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Materials Science and Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Hei Sook Sul
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kaoru Saijo
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Sanjay Kumar
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Andreas Stahl
- Program for Metabolic Biology, Department of Nutritional Sciences and Toxicology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
44
|
Abstract
Long considered inert fat storage depots, it has become clear that lipid droplets (LDs) are bona fide organelles. Like other organelles, they have a characteristic complement of proteins and lipids, and undergo a life cycle that includes biogenesis, maturation, interactions with other organelles, and turnover. I will discuss recent insights into mechanisms governing the life cycle of LDs, and compare and contrast the LD life cycle with that of other metabolic organelles such as mitochondria, peroxisomes, and autophagosomes, highlighting open questions in the field.
Collapse
Affiliation(s)
- Sarah Cohen
- University of North Carolina, Chapel Hill, NC, United States.
| |
Collapse
|
45
|
Welte MA, Gould AP. Lipid droplet functions beyond energy storage. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1260-1272. [PMID: 28735096 PMCID: PMC5595650 DOI: 10.1016/j.bbalip.2017.07.006] [Citation(s) in RCA: 364] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 02/07/2023]
Abstract
Lipid droplets are cytoplasmic organelles that store neutral lipids and are critically important for energy metabolism. Their function in energy storage is firmly established and increasingly well characterized. However, emerging evidence indicates that lipid droplets also play important and diverse roles in the cellular handling of lipids and proteins that may not be directly related to energy homeostasis. Lipid handling roles of droplets include the storage of hydrophobic vitamin and signaling precursors, and the management of endoplasmic reticulum and oxidative stress. Roles of lipid droplets in protein handling encompass functions in the maturation, storage, and turnover of cellular and viral polypeptides. Other potential roles of lipid droplets may be connected with their intracellular motility and, in some cases, their nuclear localization. This diversity highlights that lipid droplets are very adaptable organelles, performing different functions in different biological contexts. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY, United States.
| | | |
Collapse
|
46
|
Schuldiner M, Bohnert M. A different kind of love - lipid droplet contact sites. Biochim Biophys Acta Mol Cell Biol Lipids 2017. [PMID: 28627434 DOI: 10.1016/j.bbalip.2017.06.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Lipid droplets (LDs) store lipids and hence serve as energy reservoir and as a source for building-blocks for the organelle membrane systems. LD biology therefore depends on tight communication with other organelles. The unique architecture of LDs, consisting of a neutral lipid core shielded by a phospholipid-monolayer, is however an obstacle to bulk-exchange of bilayer-bounded vesicles with other organelles. In recent years, it is emerging that contact sites, places where two organelles are positioned in close proximity allowing vesicle-independent communication, are an important way to integrate LDs into the organellar landscape. However, few LD contact sites have been studied in depth and our understanding of their structure, extent and function is only starting to emerge. Here, we highlight recent findings on the functions of LD contact sites and on the proteins involved in their formation and hypothesize about the unique characteristics of the contact sites formed by these intriguing organelles. This article is part of a Special Issue entitled: Recent Advances in Lipid Droplet Biology edited by Rosalind Coleman and Matthijs Hesselink.
Collapse
Affiliation(s)
- Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maria Bohnert
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|