1
|
Hasegawa K, Fujimori H, Nakatani K, Takahashi M, Izumi Y, Bamba T, Nakamura-Shima M, Shibuya-Takahashi R, Mochizuki M, Wakui Y, Abue M, Iwai W, Fukushi D, Satoh K, Yamaguchi K, Shindo N, Yasuda J, Asano N, Imai T, Asada Y, Katori Y, Tamai K. Delta-6 desaturase FADS2 is a tumor-promoting factor in cholangiocarcinoma. Cancer Sci 2024; 115:3346-3357. [PMID: 39113435 PMCID: PMC11447924 DOI: 10.1111/cas.16306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/17/2024] [Accepted: 07/25/2024] [Indexed: 08/09/2024] Open
Abstract
Cholangiocarcinoma is a fatal disease with limited therapeutic options. We screened genes required for cholangiocarcinoma tumorigenicity and identified FADS2, a delta-6 desaturase. FADS2 depletion reduced in vivo tumorigenicity and cell proliferation. In clinical samples, FADS2 was expressed in cancer cells but not in stromal cells. FADS2 inhibition also reduced the migration and sphere-forming ability of cells and increased apoptotic cell death and ferroptosis markers. Lipidome assay revealed that triglyceride and cholesterol ester levels were decreased in FADS2-knockdown cells. The oxygen consumption ratio was also decreased in FADS2-depleted cells. These data indicate that FADS2 depletion causes a reduction in lipid levels, resulting in decrease of energy production and attenuation of cancer cell malignancy.
Collapse
Affiliation(s)
- Kohsei Hasegawa
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
- Department of Head and Neck Surgery, Miyagi Cancer Center, Natori, Miyagi, Japan
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Haruna Fujimori
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Kohta Nakatani
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Mao Nakamura-Shima
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Rie Shibuya-Takahashi
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Mai Mochizuki
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Yuta Wakui
- Division of Gastroenterology, Miyagi Cancer Center, Natori, Miyagi, Japan
| | - Makoto Abue
- Division of Gastroenterology, Miyagi Cancer Center, Natori, Miyagi, Japan
| | - Wataru Iwai
- Division of Gastroenterology, Miyagi Cancer Center, Natori, Miyagi, Japan
| | - Daisuke Fukushi
- Division of Gastroenterology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Kennichi Satoh
- Division of Gastroenterology, Tohoku Medical and Pharmaceutical University, Sendai, Miyagi, Japan
| | - Kazunori Yamaguchi
- Division of Molecular and Cellular Oncology, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Norihisa Shindo
- Division of Cancer Chromosome Biology Unit, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Jun Yasuda
- Division of Molecular and Cellular Oncology, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| | - Naoki Asano
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
- Division of Gastroenterology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takayuki Imai
- Department of Head and Neck Surgery, Miyagi Cancer Center, Natori, Miyagi, Japan
| | - Yukinori Asada
- Department of Head and Neck Surgery, Miyagi Cancer Center, Natori, Miyagi, Japan
| | - Yukio Katori
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Keiichi Tamai
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Natori, Miyagi, Japan
| |
Collapse
|
2
|
Yuan S, Zhao E. Recent advances of lipid droplet-targeted AIE-active materials for imaging, diagnosis and therapy. Biosens Bioelectron 2024; 267:116802. [PMID: 39332250 DOI: 10.1016/j.bios.2024.116802] [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: 07/22/2024] [Revised: 08/25/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
Lipid droplets (LDs) are cellular organelles specialized in the storage and regulating the release of lipids critical for energy metabolism. As investigation on LDs deepens, the complex biological functions of LDs are revealed and their relationships with various diseases such as atherosclerosis, fatty liver, obesity, and cancer are uncovered. Fluorescence-based techniques with simple operations, visible results and high non-invasiveness are ideal tools for investigating LD-related biological processes and diseases. Materials with aggregation-induced emission (AIE) characteristics have emerged as promising candidates for investigating LDs due to their high signal-to-noise ratio (S/N), strong photostability, and large Stokes shift. This review discusses the principles and advantages of LD-targeting AIE probes for imaging LDs, diagnosis of LD-associated diseases including atherosclerotic plaques, liver diseases, acute kidney diseases and cancer, therapies with LD-targeting AIE-active photosensitizers and other relevant fields in the past five years. Through typical examples, we illustrate the status of investigating LD-related imaging, diagnosis of diseases and therapy with AIE materials. This review is expected to attract attentions from scientists with different research backgrounds and contribute to the further development of LD-targeting AIE materials.
Collapse
Affiliation(s)
- Sisi Yuan
- School of Science, Harbin Institute of Technology, Shenzhen, Guangdong, 518055, China
| | - Engui Zhao
- School of Science, Harbin Institute of Technology, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
3
|
Xu C, Luo J, Yu Y. A fluorescence lifetime-based novel method for accurate lipid quantification of BODIPY vital-stained C. elegans. J Lipid Res 2024; 65:100646. [PMID: 39303981 DOI: 10.1016/j.jlr.2024.100646] [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: 07/13/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
Lipid droplets (LDs) are organelles associated with lipid storage and energy metabolism, thus, their morphology and quantity are of significant research interest. While commercially available BODIPY dye effectively labels LDs in various cell types, it also labels lysosome-related organelles (LROs) in C. elegans, leading to non-specific LD quantification. Here, we report that the fluorescent signals of BODIPY exhibit distinct fluorescence lifetime patterns for LROs and LDs, which can be captured, visualized, and filtered by fluorescence lifetime imaging microscopy. Furthermore, we proposed and validated a method based on fluorescence lifetime that can improve the accuracy of fat storage quantification in BODIPY vital-staining worms, which holds broad applications, including rapid and accurate LD quantification in forward genetic screening. Additionally, our method enables observing dynamic LD-LRO interactions in living worms, a unique capability of BODIPY vital staining. Our findings highlight distinct BODIPY fluorescence lifetime characteristics of LDs and LROs, providing a valuable tool for future research on LDs, LROs, or their interactions.
Collapse
Affiliation(s)
- Chen Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Jintao Luo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Yong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
4
|
Blume RY, Rabokon AM, Pydiura M, Yemets AI, Pirko YV, Blume YB. Genome-wide identification and evolution of the tubulin gene family in Camelina sativa. BMC Genomics 2024; 25:599. [PMID: 38877397 PMCID: PMC11177405 DOI: 10.1186/s12864-024-10503-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Tubulins play crucial roles in numerous fundamental processes of plant development. In flowering plants, tubulins are grouped into α-, β- and γ-subfamilies, while α- and β-tubulins possess a large isotype diversity and gene number variations among different species. This circumstance leads to insufficient recognition of orthologous isotypes and significantly complicates extrapolation of obtained experimental results, and brings difficulties for the identification of particular tubulin isotype function. The aim of this research is to identify and characterize tubulins of an emerging biofuel crop Camelina sativa. RESULTS We report comprehensive identification and characterization of tubulin gene family in C. sativa, including analyses of exon-intron organization, duplicated genes comparison, proper isotype designation, phylogenetic analysis, and expression patterns in different tissues. 17 α-, 34 β- and 6 γ-tubulin genes were identified and assigned to a particular isotype. Recognition of orthologous tubulin isotypes was cross-referred, involving data of phylogeny, synteny analyses and genes allocation on reconstructed genomic blocks of Ancestral Crucifer Karyotype. An investigation of expression patterns of tubulin homeologs revealed the predominant role of N6 (A) and N7 (B) subgenomes in tubulin expression at various developmental stages, contrarily to general the dominance of transcripts of H7 (C) subgenome. CONCLUSIONS For the first time a complete set of tubulin gene family members was identified and characterized for allohexaploid C. sativa species. The study demonstrates the comprehensive approach of precise inferring gene orthology. The applied technique allowed not only identifying C. sativa tubulin orthologs in model Arabidopsis species and tracking tubulin gene evolution, but also uncovered that A. thaliana is missing orthologs for several particular isotypes of α- and β-tubulins.
Collapse
Affiliation(s)
- Rostyslav Y Blume
- Institute of Food Biotechnology and Genomics of National Academy of Sciences of Ukraine, Kyiv, 02000, Ukraine.
| | - Anastasiia M Rabokon
- Institute of Food Biotechnology and Genomics of National Academy of Sciences of Ukraine, Kyiv, 02000, Ukraine
| | - Mykola Pydiura
- Institute of Food Biotechnology and Genomics of National Academy of Sciences of Ukraine, Kyiv, 02000, Ukraine
- JSC "Farmak", Kyiv, 04080, Ukraine
| | - Alla I Yemets
- Institute of Food Biotechnology and Genomics of National Academy of Sciences of Ukraine, Kyiv, 02000, Ukraine
| | - Yaroslav V Pirko
- Institute of Food Biotechnology and Genomics of National Academy of Sciences of Ukraine, Kyiv, 02000, Ukraine
| | - Yaroslav B Blume
- Institute of Food Biotechnology and Genomics of National Academy of Sciences of Ukraine, Kyiv, 02000, Ukraine
| |
Collapse
|
5
|
Maruyama R, Kudo Y, Sugiyama T. A new strategy for screening novel functional genes involved in reduction of lipid droplet accumulation. Biofactors 2024; 50:467-476. [PMID: 37983968 DOI: 10.1002/biof.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023]
Abstract
Lipid droplets (LDs) are organelles that store excess lipids and provide fatty acids for energy production during starvation. LDs are also essential for cellular maintenance, but excessive accumulation of LDs triggers various cancers in addition to metabolic diseases such as diabetes. In this study, we aimed to develop a strategy to identify new genes that reduces accumulation of LDs in cancer cells using an RNA interference (RNAi) screening system employing artificial sequence-enriched shRNA libraries. Monitoring LDs by fluorescent activated cell sorting, the subsequently collected cumulative LDs cells, and shRNA sequence analysis identified a clone that potentially functioned to accumulate LDs. The clone showed no identical sequence to human Refseq. It showed very similar sequence to seven genes by allowing three mismatches. Among these genes, we identified the mediator complex subunit 6 (MED6) gene as a target of this shRNA. Silencing of MED6 led to an increase in LD accumulation and expression of the marker genes, PLIN2 and DGAT1, in fatty cells. MED6 is a member of the mediator complex that regulates RNA polymerase II transcription through transcription factor II. Some mediator complexes play important roles in both normal and pathophysiological transcription processes. These results suggest that MED6 transcriptionally regulates the genes involved in lipid metabolism and suppresses LD accumulation.
Collapse
Affiliation(s)
- Ryuto Maruyama
- Graduate School of Bionics, Tokyo University of Technology, Tokyo, Japan
- Department of Life Science, Rikkyo University, Tokyo, Japan
| | - Yasuhiro Kudo
- Graduate School of Bionics, Tokyo University of Technology, Tokyo, Japan
| | - Tomoyasu Sugiyama
- Graduate School of Bionics, Tokyo University of Technology, Tokyo, Japan
| |
Collapse
|
6
|
Wu S, Li X, Zhou M, Cui Y, Wu W, Ping J, Guo X, Hu Q. pH-triggered hydrophility-adjustable fluorescent probes for simultaneously imaging lipid droplets and lysosomes and the application in fatty liver detection. Biosens Bioelectron 2024; 251:116084. [PMID: 38330775 DOI: 10.1016/j.bios.2024.116084] [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: 10/29/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/10/2024]
Abstract
To study the collaboration between lipid droplets (LDs) and lysosomes, and the lipid change in nonalcoholic fatty liver disease (NAFLD), herein two pH-triggered hydrophility-adjustable fluorescent probes (LD-Lyso and LD-Lyso 1) are designed. The mechanism is based on cyclization and ring-opening with thorough consideration of pH and hydrophilic differences between LDs and lysosomes. Both of the two probes exist in ring-opening form and emit red fluorescence in acidic environment, while they exist in cyclized form and the emission is blueshifted in alkaline environment due to reduced conjugate planes. Moreover, LD-Lyso exhibits near infrared fluorescence at 740 nm under ring-opening form, which facilitates further cell, tissue, and in vivo imaging. The cell imaging results show that LD-Lyso can simultaneously target LDs and lysosomes by two different colors. Impressively, LD-Lyso cannot only detect NAFLD tissues from the normal tissue, but also distinguish different degrees of NAFLD tissues and mice, which provides a very promising tool for timely diagnosis of early NAFLD.
Collapse
Affiliation(s)
- Shining Wu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250100, PR China
| | - Xuechen Li
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250100, PR China.
| | - Mingyang Zhou
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250100, PR China
| | - Yuezhi Cui
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250100, PR China
| | - Wenli Wu
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250014, PR China
| | - Jiantao Ping
- School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250014, PR China
| | - Xuezu Guo
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250100, PR China
| | - Qiongzheng Hu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250100, PR China; School of Pharmaceutical Sciences, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250014, PR China.
| |
Collapse
|
7
|
Kazemisabet F, Bahrami A, Ghosh R, Różycki B, Bahrami AH. Molecular mechanisms and energetics of lipid droplet formation and directional budding. SOFT MATTER 2024; 20:909-922. [PMID: 38189157 DOI: 10.1039/d3sm01438j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The formation and budding of lipid droplets (LDs) are known to be governed by the LD size and by membrane tensions in the endoplasmic reticulum (ER) bilayer and LD-monolayers. Using coarse-grained simulations of an LD model, we first show that ER-embedded LDs of different sizes can form through a continuous transition from wide LD lenses to spherical LDs at a fixed LD size. The ER tendency to relax its bilayer modulates the transition via a subtle interplay between the ER and LD lipid densities. By calculating the energetic landscape of the LD transition, we demonstrate that this size-independent transition is regulated by the mechanical force balance of ER and LD-tensions, independent from membrane bending and line tension whose energetic contributions are negligible according to our calculations. Our findings explain experimental observation of stable LDs of various shapes. We then propose a novel mechanism for directional LD budding where the required membrane asymmetry is provided by the exchange of lipids between the LD-monolayers. Remarkably, we demonstrate that this budding process is energetically neutral. Consequently, LD budding can proceed by a modest energy input from proteins or other driving agents. We obtain equal lipid densities and membrane tensions in LD-monolayers throughout budding. Our findings indicate that unlike LD formation, LD budding by inter-monolayer lipid exchange is a tension-independent process.
Collapse
Affiliation(s)
- Fatemeh Kazemisabet
- School of Mechanical Engineering, College of Engineering, University of Tehran, North Kargar St., 14399-57131 Tehran, Iran
| | - Arash Bahrami
- School of Mechanical Engineering, College of Engineering, University of Tehran, North Kargar St., 14399-57131 Tehran, Iran
| | - Rikhia Ghosh
- Department of Pharmacological Sciences, Icahn School of Medicine, Mount Sinai, New York 10029, USA
| | - Bartosz Różycki
- Institute of Physics, Polish Academy of Sciences, Al. Lotników 32/46, 02-668 Warsaw, Poland
| | - Amir H Bahrami
- UNAM-National Nanotechnology Research Center and Institute of Materials Science & Nanotechnology, Bilkent University, Ankara, Turkey.
- Living Matter Physics, Max Planck Institute for Dynamics and Self-Organization, 37077 Göttingen, Germany
| |
Collapse
|
8
|
Dargham T, Mallick I, Kremer L, Santucci P, Canaan S. Intrabacterial lipid inclusion-associated proteins: a core machinery conserved from saprophyte Actinobacteria to the human pathogen Mycobacterium tuberculosis. FEBS Open Bio 2023; 13:2306-2323. [PMID: 37872001 PMCID: PMC10699116 DOI: 10.1002/2211-5463.13721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/02/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb), the aetiologic agent of tuberculosis (TB), stores triacylglycerol (TAG) in the form of intrabacterial lipid inclusions (ILI) to survive and chronically persist within its host. These highly energetic molecules represent a major source of carbon to support bacterial persistence and reactivation, thus playing a leading role in TB pathogenesis. However, despite its physiological and clinical relevance, ILI metabolism in Mtb remains poorly understood. Recent discoveries have suggested that several ILI-associated proteins might be widely conserved across TAG-producing prokaryotes, but still very little is known regarding the nature and the biological functions of these proteins. Herein, we performed an in silico analysis of three independent ILI-associated proteomes previously reported to computationally define a potential core ILI-associated proteome, referred to as ILIome. Our investigation revealed the presence of 70 orthologous proteins that were strictly conserved, thereby defining a minimal ILIome core. We further narrowed our analysis to proteins involved in lipid metabolism and discuss here their putative biological functions, along with their molecular interactions and dynamics at the surface of these bacterial organelles. We also highlight the experimental limitations of the original proteomic investigations and of the present bioinformatic analysis, while describing new technological approaches and presenting biological perspectives in the field. The in silico investigation presented here aims at providing useful datasets that could constitute a scientific resource of broad interest for the mycobacterial community, with the ultimate goal of enlightening ILI metabolism in prokaryotes with a special emphasis on Mtb pathogenesis.
Collapse
Affiliation(s)
- Tonia Dargham
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
- IHU Méditerranée InfectionAix‐Marseille Univ.France
| | - Ivy Mallick
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
| | - Laurent Kremer
- Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche en Infectiologie de Montpellier (IRIM)Université de MontpellierFrance
- INSERM, Institut de Recherche en Infectiologie de MontpellierFrance
| | - Pierre Santucci
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
| | - Stéphane Canaan
- Aix‐Marseille Univ, CNRS, LISM UMR 7255, IMM FR3479, IM2BFrance
| |
Collapse
|
9
|
Cao Z, Zhang Q, Zhou Z, Xu S, Pan B, Zhang S, Zhang G, Zhi Z, Shi Y, Cui L, Liu P. Construction and application of artificial lipoproteins using adiposomes. J Lipid Res 2023; 64:100436. [PMID: 37648212 PMCID: PMC10518588 DOI: 10.1016/j.jlr.2023.100436] [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: 07/24/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
Lipoproteins are complex particles comprised of a neutral lipid core wrapped with a phospholipid monolayer membrane and apolipoproteins on the membrane, which is closely associated with metabolic diseases. To facilitate the elucidation of its formation and dynamics, as well as its applications, we developed an in vitro system in which adiposomes, consisting of a hydrophobic core encircled by a monolayer-phospholipid membrane, were engineered into artificial lipoproteins (ALPs) by recruiting one or more kinds of apolipoproteins, for example, apolipoprotein (Apo) A-I, ApoE, ApoA-IV, and ApoB. In vitro and in vivo studies demonstrated the stability and biological activity of ALPs derived from adiposomes, which resembles native lipoproteins. Of note, adiposomes bearing ApoE were internalized via clathrin-mediated endocytosis following LDLR binding and were delivered to lysosomes. On the other hand, adiposomes bearing ApoA-IV mimicked the existing form of endogenous ApoA-IV and exhibited significant improvement in glucose tolerance in mice. In addition, the construction process was simple, precise, reproducible, as well as easy to adjust for mass production. With this experimental system, different apolipoproteins can be recruited to build ALPs for some biological goals and potential applications in biomedicine.
Collapse
Affiliation(s)
- Zhen Cao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Qi Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Ziyun Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Bin Pan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shuyan Zhang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Infectious Diseases, Beijing, China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing, China
| | - Gaoxin Zhang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, China
| | - Zelun Zhi
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Yumeng Shi
- College of Life Science, Hebei Normal University, Shijiazhuang, China
| | - Liujuan Cui
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
10
|
Yang Y, Luo Y, Yi S, Gao Q, Gong T, Feng Y, Wu D, Zheng X, Wang H, Zhang G, Sun Y. Porcine reproductive and respiratory syndrome virus regulates lipid droplet accumulation in lipid metabolic pathways to promote viral replication. Virus Res 2023; 333:199139. [PMID: 37217033 PMCID: PMC10352717 DOI: 10.1016/j.virusres.2023.199139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 05/24/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is a severe respiratory disease caused by porcine reproductive and respiratory syndrome virus (PRRSV) that can lead to the abortion of pregnant sows and decreased boar semen quality. However, the mechanisms of PRRSV replication in the host have not yet been fully elucidated. As lipid metabolism and lipid droplets (LDs) have been reported to play important roles in the replication of various viruses, we aimed to explore the mechanisms through which LDs affect PRRSV replication. Laser confocal and transmission electron microscopy revealed that PRRSV infection promoted intracellular LD accumulation, which was significantly reduced by treatment with the NF-κB signaling pathway inhibitors BAY11-7082 and metformin hydrochloride (MH). In addition, treatment with a DGAT1 inhibitor significantly reduced the protein expression of Phosphorylated NF-ΚB P65and PIκB and the transcription of IL-1β and IL-8 in the NF-κB signaling pathway. Furthermore, we showed that the reduction of the NF-κB signaling pathway and LDs significantly reduced PRRSV replication. Together, the findings of this study suggest a novel mechanism through which PRRSV regulates the NF-κB signaling pathway to increase LD accumulation and promote viral replication. Moreover, we demonstrated that both BAY11-7082 and MH can reduce PRRSV replication by reducing the NF-κB signaling pathway and LD accumulation. This study lays a theoretical foundation for research on the mechanism of PRRS prevention and control, as well as the research and development of antiviral drugs.
Collapse
Affiliation(s)
- Yunlong Yang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Yizhuo Luo
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Songqiang Yi
- Agricultural Technology Extension Center of Jiangxi Province, Nanchang, China
| | - Qi Gao
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Ting Gong
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, PR China
| | - Yongzhi Feng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Dongdong Wu
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, PR China
| | - Xiaoyu Zheng
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, PR China
| | - Heng Wang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China
| | - Guihong Zhang
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, PR China
| | - Yankuo Sun
- Guangdong Provincial Key Laboratory of Zoonosis Prevention and Control, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China; Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Maoming 525000, China; Key Laboratory of Animal Vaccine Development, Ministry of Agriculture and Rural Affairs, PR China.
| |
Collapse
|
11
|
Sun Y, Heng J, Liu F, Zhang S, Liu P. Isolation and proteomic study of fish liver lipid droplets. BIOPHYSICS REPORTS 2023; 9:120-133. [PMID: 38028150 PMCID: PMC10648235 DOI: 10.52601/bpr.2023.230004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/02/2023] [Indexed: 12/01/2023] Open
Abstract
Lipid droplets (LDs) are a neutral lipid storage organelle that is conserved in almost all species. Excessive storage of neutral lipids in LDs is directly associated with many metabolic syndromes. Zebrafish is a better model animal for the study of LD biology due to its transparent embryonic stage compared to other organisms. However, the study of LDs in fish has been difficult due to the lack of specific LD marker proteins and the limitation of purification technology. In this paper, the purification and proteomic analysis of liver LDs of fish including zebrafish and Carassius auratus were performed for the first time. 259 and 267 proteins were identified respectively. Besides most of the identified proteins were reported in previous LD proteomes of mammals, indicating the similarity between mammal and fish LDs. We also identified many unique proteins of liver LDs in fish that are involved in the regulation of LD dynamics. Through morphological and biochemical analysis, we found that the marker protein Plin2 of zebrafish LD was located on LDs in Huh7 cells. These results will facilitate further study of LDs in fish and liver metabolic diseases using fish as a model animal.
Collapse
Affiliation(s)
- Yuwei Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Heng
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Institute of Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
12
|
Nakamura E, Maekawa K, Saito Y, Matsumoto T, Ogawa M, Komohara Y, Asada Y, Yamashita A. Altered choline level in atherosclerotic lesions: Upregulation of choline transporter-like protein 1 in human coronary unstable plaque. PLoS One 2023; 18:e0281730. [PMID: 36800352 PMCID: PMC9937458 DOI: 10.1371/journal.pone.0281730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Inflammatory activity and hypoxia in atherosclerotic plaques are associated with plaque instability and thrombotic complications. Recent studies show that vascular cell metabolism affects atherogenesis and thrombogenicity. This study aimed to identify the metabolites in macrophage-rich unstable plaques that modulate atherogenesis and serve as potential markers of plaque instability. Atherosclerotic plaques were induced by balloon injury in the iliofemoral arteries of rabbits fed on a conventional or 0.5% cholesterol diet. At 3 months post-balloon injury, the arteries and cardiac tissues were subjected to histological, quantitative real-time polymerase chain reaction, and metabolomic analyses. The identified metabolite-related proteins were immunohistochemically analyzed in stable and unstable plaques from human coronary arteries. The factors modulating the identified metabolites were examined in macrophages derived from human peripheral blood mononuclear cells. Metabolomic analysis revealed that choline and guanine levels in macrophage-rich arteries were upregulated compared with those in non-injured arteries and cardiac tissues. Vascular choline levels, but not guanine levels, were positively correlated with the areas immunopositive for macrophages and tumor necrosis factor (TNF)-α and matrix metalloproteinase (MMP) 9 mRNA levels in injured arteries. In human coronary arteries, choline transporter-like protein (CTL) 1 was mainly localized to macrophages within plaques. The area that was immunopositive for CTL1 in unstable plaques was significantly higher than that in stable plaques. Intracellular choline levels were upregulated upon stimulation with TNF-α but were downregulated under hypoxia in cultured macrophages. Administration of choline upregulated the expression of TNF-α and CTL1 mRNA in cultured macrophages. The transfection of CTL1 small interfering RNA decreased CTL1, TNF-α, and MMP9 mRNA levels in cultured macrophages. These results suggest that choline metabolism is altered in macrophage-rich atherosclerotic lesions and unstable plaques. Thus, CTL1 may be potential markers of plaque instability.
Collapse
Affiliation(s)
- Eriko Nakamura
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazunari Maekawa
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yoichi Saito
- Bioengineering Lab, Faculty of Advanced Science and Technology, Kumamoto University, Kumamoto, Japan
| | - Tomoko Matsumoto
- Center for Collaborative Research and Community Cooperation, University of Miyazaki, Miyazaki, Japan
| | - Mikako Ogawa
- Laboratory of Bioanalysis and Molecular Imaging, Graduate School of Pharmaceutical Science, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoshihiro Komohara
- Department of Cell Pathology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yujiro Asada
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- Department of Pathology, Miyazaki Medical Association Hospital, Miyazaki, Japan
| | - Atsushi Yamashita
- Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
- * E-mail:
| |
Collapse
|
13
|
Fang X, Zhou Y, Chen S, Xu X, Ke J, Zhou Y, Wei H, Fu B. Natural killer cells promote intra-cellular-infected trophoblasts survival via APOD-LRP1 axis. Immunology 2022. [PMID: 36562137 DOI: 10.1111/imm.13621] [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: 10/03/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Natural killer (NK) cells are known for their potent ability to kill stressed cells, whereas host cells infected with intra-cellular bacteria may also be benefit from the selective killing function of NK cells and survive. The mechanism of how NK cells protect host cells infected with intra-cellular bacteria is still unclear. Here, we discovered that decidual NK (dNK) cells cannot only eliminate intra-cellular bacteria which infected trophoblasts, but can also synthesize more lipids and transport lipids to trophoblasts to avoid their apoptosis. Mechanically, NK cells synthesize more lipids accompanied by increasing expression of apolipoprotein APOD. Lipids in NK cells can be delivered to trophoblast cells through APOD, maintaining adequate lipid droplet content and lipid metabolism homeostasis in trophoblasts. Blocking the APOD receptor LRP1 abolished lipid transport from NK cells to trophoblasts, and the reduction of lipid droplets caused by bacterial infection in trophoblast cells could not be restored, culminating in cell apoptosis. Our study provides new evidence for the immune surveillance and protective effect of NK cells on embryos during early pregnancy.
Collapse
Affiliation(s)
- Xi Fang
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Yonggang Zhou
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Siao Chen
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Xiuxiu Xu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Jieqi Ke
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Haiming Wei
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Binqing Fu
- The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Immunology, University of Science and Technology of China, Hefei, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
14
|
Singh J, Sanghavi P, Mallik R. Microtubule motor driven interactions of lipid droplets: Specificities and opportunities. Front Cell Dev Biol 2022; 10:893375. [PMID: 36200039 PMCID: PMC9527339 DOI: 10.3389/fcell.2022.893375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Lipid Droplets (LDs) are evolutionarily conserved cellular organelles that store neutral lipids such as triacylglycerol and cholesterol-esters. Neutral lipids are enclosed within the limiting membrane of the LD, which is a monolayer of phospholipids and is therefore fundamentally different from the bilayer membrane enclosing most other organelles. LDs have long been viewed as a storehouse of lipids needed on demand for generating energy and membranes inside cells. Outside this classical view, we are now realizing that LDs have significant roles in protein sequestration, supply of signalling lipids, viral replication, lipoprotein production and many other functions of important physiological consequence. To execute such functions, LDs must often exchange lipids and proteins with other organelles (e.g., the ER, lysosomes, mitochondria) via physical contacts. But before such exchanges can occur, how does a micron-sized LD with limited ability to diffuse around find its cognate organelle? There is growing evidence that motor protein driven motion of LDs along microtubules may facilitate such LD-organelle interactions. We will summarize some aspects of LD motion leading to LD-organelle contacts, how these change with metabolic state and pathogen infections, and also ask how these pathways could perhaps be targeted selectively in the context of disease and drug delivery. Such a possibility arises because the binding of motor proteins to the monolayer membrane on LDs could be different from motor binding to the membrane on other cellular organelles.
Collapse
Affiliation(s)
- Jagjeet Singh
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
- *Correspondence: Roop Mallik, ; Jagjeet Singh,
| | - Paulomi Sanghavi
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, India
| | - Roop Mallik
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
- *Correspondence: Roop Mallik, ; Jagjeet Singh,
| |
Collapse
|
15
|
Yang R, Yi M, Xiang B. Novel Insights on Lipid Metabolism Alterations in Drug Resistance in Cancer. Front Cell Dev Biol 2022; 10:875318. [PMID: 35646898 PMCID: PMC9136290 DOI: 10.3389/fcell.2022.875318] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/13/2022] [Indexed: 12/26/2022] Open
Abstract
Chemotherapy is one of the primary treatments for most human cancers. Despite great progress in cancer therapeutics, chemotherapy continues to be important for improving the survival of cancer patients, especially for those who has unresectable metastatic tumors or fail to respond to immunotherapy. However, intrinsic or acquired chemoresistance results in tumor recurrence, which remains a major obstacle in anti-cancer treatment. The high prevalence of chemoresistant cancer makes it urgent to deepen our understanding on chemoresistance mechanisms and to develop novel therapeutic strategies. Multiple mechanisms, including drug efflux, enhanced DNA damage reparability, increased detoxifying enzymes levels, presence of cancer stem cells (CSCs), epithelial mesenchymal transition (EMT), autophagy, ferroptosis and resistance to apoptosis, underlie the development of chemoresistance. Recently, accumulating evidence suggests that lipid metabolism alteration is closely related to drug resistance in tumor. Targeting lipid metabolism in combination with traditional chemotherapeutic drugs is a promising strategy to overcome drug resistance. Therefore, this review compiles the current knowledge about aberrant lipid metabolism in chemoresistant cancer, mainly focusing on aberrant fatty acid metabolism, and presents novel therapeutic strategies targeting altered lipid metabolism to overcome chemoresistance in cancer.
Collapse
Affiliation(s)
- Ruixue Yang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hypertension Center, FuWai Hospital, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mei Yi
- Department of Dermatology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Bo Xiang
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
- The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
16
|
Ahirwar A, Kesharwani K, Deka R, Muthukumar S, Khan MJ, Rai A, Vinayak V, Varjani S, Joshi KB, Morjaria S. Microalgal drugs: A promising therapeutic reserve for the future. J Biotechnol 2022; 349:32-46. [PMID: 35339574 DOI: 10.1016/j.jbiotec.2022.03.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/17/2022] [Accepted: 03/20/2022] [Indexed: 12/16/2022]
Abstract
Over the decades, a variety of chemically synthesized drugs are being used to cure existing diseases but often these drugs could not be effectively employed for the treatment of serious and newly emerging diseases. Fortunately, in nature there occurs immense treasure of plants and microorganisms which are living jewels with respect to their richness of medically important metabolites of high value. Hence, amongst the existing microorganism(s), the marine world offers a plethora of biological entities that can contribute to alleviate numerous human ailments. Algae are one such photosynthetic microorganism found in both marine as well as fresh water which are rich source of metabolites known for their nutrient content and health benefits. Various algal species like Haematococcus, Diatoms, Griffithsia, Chlorella, Spirulina, Ulva, etc. have been identified and isolated to produce biologically active and pharmaceutically important high value compounds like astaxanthin, fucoxanthin, sulphur polysaccharides mainly galactose, rhamnose, xylose, fucose etc., which show antimicrobial, antifungal, anti-cancer, and antiviral activities. However, the production of either of these bio compounds is favored under conditions of stress. This review gives detailed information on various nutraceutical metabolites extracted from algae. Additionally focus has been made on the role of these bio compounds extracted from algae especially sulphur polysaccharides to treat several diseases with prospective treatment for SARS-CoV-2. Lastly it covers the knowledge gaps and future perspectives in this area of research.
Collapse
Affiliation(s)
- Ankesh Ahirwar
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India
| | - Khushboo Kesharwani
- Department of Chemistry, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India
| | - Rahul Deka
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India
| | - Shreya Muthukumar
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India
| | - Mohd Jahir Khan
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India
| | - Anshuman Rai
- MMU, Deemed University, School of Engineering, Department of Biotechnology, Ambala, Haryana, 133203, India
| | - Vandana Vinayak
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India.
| | - Sunita Varjani
- Gujarat Pollution Control Board, Gandhinagar, Gujarat, 382 010, India.
| | - Khashti Ballabh Joshi
- Department of Chemistry, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India
| | - Shruti Morjaria
- Diatom Nanoengineering and Metabolism Laboratory (DNM), School of Applied Science, Dr. Harisingh Gour Central University, Sagar (MP) 470003, India
| |
Collapse
|
17
|
Pressly JD, Gurumani MZ, Varona Santos JT, Fornoni A, Merscher S, Al-Ali H. Adaptive and maladaptive roles of lipid droplets in health and disease. Am J Physiol Cell Physiol 2022; 322:C468-C481. [PMID: 35108119 PMCID: PMC8917915 DOI: 10.1152/ajpcell.00239.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Advances in the understanding of lipid droplet biology have revealed essential roles for these organelles in mediating proper cellular homeostasis and stress response. Lipid droplets were initially thought to play a passive role in energy storage. However, recent studies demonstrate that they have substantially broader functions, including protection from reactive oxygen species, endoplasmic reticulum stress, and lipotoxicity. Dysregulation of lipid droplet homeostasis is associated with various pathologies spanning neurological, metabolic, cardiovascular, oncological, and renal diseases. This review provides an overview of the current understanding of lipid droplet biology in both health and disease.
Collapse
Affiliation(s)
- Jeffrey D. Pressly
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Margaret Z. Gurumani
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Javier T. Varona Santos
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Alessia Fornoni
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Sandra Merscher
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida
| | - Hassan Al-Ali
- 1Katz Division of Nephrology and Hypertension and Katz Family Drug Discovery Center, University of Miami, Miller School of Medicine, Miami, Florida,2Department of Medicine, University of Miami, Miller School of Medicine, Miami, Florida,3Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, Florida,4The Miami Project to Cure Paralysis, University of Miami, Miller School of Medicine, Miami, Florida,5Sylvester Comprehensive Cancer Center, University of Miami, Miller School of Medicine, Miami, Florida
| |
Collapse
|
18
|
Fader Kaiser CM, Romano PS, Vanrell MC, Pocognoni CA, Jacob J, Caruso B, Delgui LR. Biogenesis and Breakdown of Lipid Droplets in Pathological Conditions. Front Cell Dev Biol 2022; 9:826248. [PMID: 35198567 PMCID: PMC8860030 DOI: 10.3389/fcell.2021.826248] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022] Open
Abstract
Lipid droplets (LD) have long been considered as mere fat drops; however, LD have lately been revealed to be ubiquitous, dynamic and to be present in diverse organelles in which they have a wide range of key functions. Although incompletely understood, the biogenesis of eukaryotic LD initiates with the synthesis of neutral lipids (NL) by enzymes located in the endoplasmic reticulum (ER). The accumulation of NL leads to their segregation into nanometric nuclei which then grow into lenses between the ER leaflets as they are further filled with NL. The lipid composition and interfacial tensions of both ER and the lenses modulate their shape which, together with specific ER proteins, determine the proneness of LD to bud from the ER toward the cytoplasm. The most important function of LD is the buffering of energy. But far beyond this, LD are actively integrated into physiological processes, such as lipid metabolism, control of protein homeostasis, sequestration of toxic lipid metabolic intermediates, protection from stress, and proliferation of tumours. Besides, LD may serve as platforms for pathogen replication and defense. To accomplish these functions, from biogenesis to breakdown, eukaryotic LD have developed mechanisms to travel within the cytoplasm and to establish contact with other organelles. When nutrient deprivation occurs, LD undergo breakdown (lipolysis), which begins with the LD-associated members of the perilipins family PLIN2 and PLIN3 chaperone-mediated autophagy degradation (CMA), a specific type of autophagy that selectively degrades a subset of cytosolic proteins in lysosomes. Indeed, PLINs CMA degradation is a prerequisite for further true lipolysis, which occurs via cytosolic lipases or by lysosome luminal lipases when autophagosomes engulf portions of LD and target them to lysosomes. LD play a crucial role in several pathophysiological processes. Increased accumulation of LD in non-adipose cells is commonly observed in numerous infectious diseases caused by intracellular pathogens including viral, bacterial, and parasite infections, and is gradually recognized as a prominent characteristic in a variety of cancers. This review discusses current evidence related to the modulation of LD biogenesis and breakdown caused by intracellular pathogens and cancer.
Collapse
Affiliation(s)
- Claudio M Fader Kaiser
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Patricia S Romano
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - M Cristina Vanrell
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Cristian A Pocognoni
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Julieta Jacob
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| | - Benjamín Caruso
- Instituto de Investigaciones Biologicas y Tecnologicas, Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Cordoba, Cordoba, Argentina
| | - Laura R Delgui
- CONICET Dr. Mario H. Burgos Institute of Histology and Embryology (IHEM), Mendoza, Argentina
| |
Collapse
|
19
|
Zhou X, Mo Z, Li Y, Huang L, Yu S, Ge L, Hu Y, Shi S, Zhang L, Wang L, Gao L, Yang G, Chu G. Oleic acid reduces steroidogenesis by changing the lipid type stored in lipid droplets of ovarian granulosa cells. J Anim Sci Biotechnol 2022; 13:27. [PMID: 35130983 PMCID: PMC8822748 DOI: 10.1186/s40104-021-00660-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/29/2021] [Indexed: 12/27/2022] Open
Abstract
Background Oleic acid is an abundant free fatty acid present in livestock that are in a negative energy-balance state, and it may have detrimental effects on female reproduction and fertility. Oleic acid induces lipid accumulation in bovine granulosa cells, which leads to a foam cell-like morphology and reduced steroidogenesis. However, why oleic acid increases lipid accumulation but decreases steroidogenesis remains unclear. This study focused on oleic acid’s effects on lipid type and steroidogenesis. Results Oleic acid increased the lipid accumulation in a concentration-dependent manner and mainly increased the triglyceride level and decreased the cholesterol ester level. Oleic acid also led to a decline in estradiol and progesterone production in porcine granulosa cells in vitro. In addition, oleic acid up-regulated the expression of CD36 and diacylglycerol acyltransferase 2, but down-regulated the expression of 3-hydroxy-3-methylglutaryl-coenzyme A reductase, scavenger receptor class B member 1 and acetyl-Coenzyme A acetyltransferase 2, as well as steroidogenesis-related genes, including cytochrome P450 family 11 subfamily A member 1, cytochrome P450 family 19 subfamily A member 1 and 3 as well as steroidogenic acute regulatory protein at the mRNA and protein levels. An oleic acid-rich diet also enhanced the triglyceride levels and reduced the cholesterol levels in ovarian tissues of female mice, which resulted in lower estradiol levels than in control-fed mice. Compared with the control, decreases in estrus days and the numbers of antral follicles and corpora lutea, as well as an increase in the numbers of the atretic follicles, were found in the oleic acid-fed female mice. Conclusions Oleic acid changed the lipid type stored in lipid droplets of ovarian granulosa cells, and led to a decrease in steroidogenesis. These results improve our understanding of fertility decline in livestock that are in a negative energy-balance state.
Collapse
Affiliation(s)
- Xiaoge Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Zhaoyi Mo
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yankun Li
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Liang Huang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Sihai Yu
- College of veterinary medicine, Northwest A&F University, Yangling, 712100, China
| | - Lan Ge
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yamei Hu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Shengjie Shi
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lutong Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Liguang Wang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Lei Gao
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China.,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Gongshe Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China. .,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Guiyan Chu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, Yangling, 712100, China. .,Laboratory of Animal Fat Deposition & Muscle Development, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
20
|
Zhang S, Peng X, Yang S, Li X, Huang M, Wei S, Liu J, He G, Zheng H, Yang L, Li H, Fan Q. The regulation, function, and role of lipophagy, a form of selective autophagy, in metabolic disorders. Cell Death Dis 2022; 13:132. [PMID: 35136038 PMCID: PMC8825858 DOI: 10.1038/s41419-022-04593-3] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/07/2022] [Accepted: 01/27/2022] [Indexed: 12/15/2022]
Abstract
Autophagy is a conserved method of quality control in which cytoplasmic contents are degraded via lysosomes. Lipophagy, a form of selective autophagy and a novel type of lipid metabolism, has recently received much attention. Lipophagy is defined as the autophagic degradation of intracellular lipid droplets (LDs). Although much remains unknown, lipophagy appears to play a significant role in many organisms, cell types, metabolic states, and diseases. It participates in the regulation of intracellular lipid storage, intracellular free lipid levels (e.g., fatty acids), and energy balance. However, it remains unclear how intracellular lipids regulate autophagy. Impaired lipophagy can cause cells to become sensitive to death stimuli and may be responsible for the onset of a variety of diseases, including nonalcoholic fatty liver disease and metabolic syndrome. Like autophagy, the role of lipophagy in cancer is poorly understood, although analysis of specific autophagy receptors has helped to expand the diversity of chemotherapeutic targets. These studies have stimulated increasing interest in the role of lipophagy in the pathogenesis and treatment of cancer and other human diseases.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Mingyao Huang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Shibo Wei
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Jiaxing Liu
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Guangpeng He
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hongyu Zheng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Qing Fan
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
21
|
Chatterjee R, Chowdhury AR, Mukherjee D, Chakravortty D. Lipid larceny: channelizing host lipids for establishing successful pathogenesis by bacteria. Virulence 2021; 12:195-216. [PMID: 33356849 PMCID: PMC7808437 DOI: 10.1080/21505594.2020.1869441] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 12/03/2020] [Accepted: 12/17/2020] [Indexed: 12/16/2022] Open
Abstract
Lipids are complex organic compounds made up of carbon, oxygen, and hydrogen. These play a diverse and intricate role in cellular processes like membrane trafficking, protein sorting, signal transduction, and bacterial infections. Both Gram-positive bacteria (Staphylococcus sp., Listeria monocytogenes, etc.) and Gram-negative bacteria (Chlamydia sp., Salmonella sp., E. coli, etc.) can hijack the various host-lipids and utilize them structurally as well as functionally to mount a successful infection. The pathogens can deploy with various arsenals to exploit host membrane lipids and lipid-associated receptors as an attachment for toxins' landing or facilitate their entry into the host cellular niche. Bacterial species like Mycobacterium sp. can also modulate the host lipid metabolism to fetch its carbon source from the host. The sequential conversion of host membrane lipids into arachidonic acid and prostaglandin E2 due to increased activity of cPLA-2 and COX-2 upon bacterial infection creates immunosuppressive conditions and facilitates the intracellular growth and proliferation of bacteria. However, lipids' more debatable role is that they can also be a blessing in disguise. Certain host-lipids, especially sphingolipids, have been shown to play a crucial antibacterial role and help the host in combating the infections. This review shed light on the detailed role of host lipids in bacterial infections and the current understanding of the lipid in therapeutics. We have also discussed potential prospects and the need of the hour to help us cope in this race against deadly pathogens and their rapidly evolving stealthy virulence strategies.
Collapse
Affiliation(s)
- Ritika Chatterjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Atish Roy Chowdhury
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Debapriya Mukherjee
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
| | - Dipshikha Chakravortty
- Department of Microbiology and Cell Biology, Division of Biological Sciences, Indian Institute of Science, Bangalore, India
- Centre for Biosystems Science and Engineering, Indian Institute of Science, Bangalore, India
| |
Collapse
|
22
|
Lipids in Pathophysiology and Development of the Membrane Lipid Therapy: New Bioactive Lipids. MEMBRANES 2021; 11:membranes11120919. [PMID: 34940418 PMCID: PMC8708953 DOI: 10.3390/membranes11120919] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 12/19/2022]
Abstract
Membranes are mainly composed of a lipid bilayer and proteins, constituting a checkpoint for the entry and passage of signals and other molecules. Their composition can be modulated by diet, pathophysiological processes, and nutritional/pharmaceutical interventions. In addition to their use as an energy source, lipids have important structural and functional roles, e.g., fatty acyl moieties in phospholipids have distinct impacts on human health depending on their saturation, carbon length, and isometry. These and other membrane lipids have quite specific effects on the lipid bilayer structure, which regulates the interaction with signaling proteins. Alterations to lipids have been associated with important diseases, and, consequently, normalization of these alterations or regulatory interventions that control membrane lipid composition have therapeutic potential. This approach, termed membrane lipid therapy or membrane lipid replacement, has emerged as a novel technology platform for nutraceutical interventions and drug discovery. Several clinical trials and therapeutic products have validated this technology based on the understanding of membrane structure and function. The present review analyzes the molecular basis of this innovative approach, describing how membrane lipid composition and structure affects protein-lipid interactions, cell signaling, disease, and therapy (e.g., fatigue and cardiovascular, neurodegenerative, tumor, infectious diseases).
Collapse
|
23
|
Genetic engineering of microalgae for enhanced lipid production. Biotechnol Adv 2021; 52:107836. [PMID: 34534633 DOI: 10.1016/j.biotechadv.2021.107836] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022]
Abstract
Microalgae have the potential to become microbial cell factories for lipid production. Their ability to convert sunlight and CO2 into valuable lipid compounds has attracted interest from cosmetic, biofuel, food and feed industries. In order to make microalgae-derived products cost-effective and commercially competitive, enhanced growth rates and lipid productivities are needed, which require optimization of cultivation systems and strain improvement. Advances in genetic tool development and omics technologies have increased our understanding of lipid metabolism, which has opened up possibilities for targeted metabolic engineering. In this review we provide a comprehensive overview on the developments made to genetically engineer microalgal strains over the last 30 years. We focus on the strategies that lead to an increased lipid content and altered fatty acid profile. These include the genetic engineering of the fatty acid synthesis pathway, Kennedy pathway, polyunsaturated fatty acid and triacylglycerol metabolisms and fatty acid catabolism. Moreover, genetic engineering of specific transcription factors, NADPH generation and central carbon metabolism, which lead to increase of lipid accumulation are also reviewed.
Collapse
|
24
|
Lipid droplets are beneficial for rabies virus replication by facilitating viral budding. J Virol 2021; 96:e0147321. [PMID: 34757839 DOI: 10.1128/jvi.01473-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rabies is an old zoonotic disease caused by rabies virus (RABV), but the pathogenic mechanism of RABV is still not completely understood. Lipid droplets have been reported to play a role in pathogenesis of several viruses. However, its role on RABV infection remains unclear. Here, we initially found that RABV infection upregulated lipid droplet (LD) production in multiple cells and mouse brains. After the treatment of atorvastatin, a specific inhibitor of LD, RABV replication in N2a cells decreased. Then we found that RABV infection could upregulate N-myc downstream regulated gene-1 (NDRG1), which in turn enhance the expression of diacylglycerol acyltransferase 1/2 (DGAT1/2). DGAT1/2 could elevate cellular triglycerides synthesis and ultimately promote intracellular LD formation. Furthermore, we found that RABV-M and RABV-G, which were mainly involved in the viral budding process, could colocalize with LDs, indicating that RABV might utilize LDs as a carrier to facilitate viral budding and eventually increase virus production. Taken together, our study reveals that lipid droplets are beneficial for RABV replication and their biogenesis is regulated via NDRG1-DGAT1/2 pathway, which provides novel potential targets for developing anti-RABV drugs. IMPORTANCE Lipid droplets have been proven to play an important role in viral infections, but its role in RABV infection has not yet been elaborated. Here, we find that RABV infection upregulates the generation of LDs by enhancing the expression of N-myc downstream regulated gene-1 (NDRG1). Then NDRG1 elevated cellular triglycerides synthesis by increasing the activity of diacylglycerol acyltransferase 1/2 (DGAT1/2), which promotes the biogenesis of LDs. RABV-M and RABV-G, which are the major proteins involved in viral budding, could utilize LDs as a carrier and transport to cell membrane, resulting in enhanced virus budding. Our findings will extend the knowledge of lipid metabolism in RABV infection and help to explore potential therapeutic targets for RABV.
Collapse
|
25
|
Zhang J, Hu Y, Wang Y, Fu L, Xu X, Li C, Xu J, Li C, Zhang L, Yang R, Jiang X, Wu Y, Liu P, Zou X, Liang B. mmBCFA C17iso ensures endoplasmic reticulum integrity for lipid droplet growth. J Cell Biol 2021; 220:212690. [PMID: 34623380 PMCID: PMC8563294 DOI: 10.1083/jcb.202102122] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 08/22/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
In eukaryote cells, lipid droplets (LDs) are key intracellular organelles that dynamically regulate cellular energy homeostasis. LDs originate from the ER and continuously contact the ER during their growth. How the ER affects LD growth is largely unknown. Here, we show that RNAi knockdown of acs-1, encoding an acyl-CoA synthetase required for the biosynthesis of monomethyl branched-chain fatty acids C15iso and C17iso, remarkably prevented LD growth in Caenorhabditis elegans. Dietary C17iso, or complex lipids with C17iso including phosphatidylcholine, phosphatidylethanolamine, and triacylglycerol, could fully restore the LD growth in the acs-1RNAi worms. Mechanistically, C17iso may incorporate into phospholipids to ensure the membrane integrity of the ER so as to maintain the function of ER-resident enzymes such as SCD/stearoyl-CoA desaturase and DGAT2/diacylglycerol acyltransferase for appropriate lipid synthesis and LD growth. Collectively, our work uncovers a unique fatty acid, C17iso, as the side chain of phospholipids for determining the ER homeostasis for LD growth in an intact organism, C. elegans.
Collapse
Affiliation(s)
- Jingjing Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Ying Hu
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yanli Wang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Lin Fu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xiumei Xu
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China.,Institute of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
| | - Chunxia Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China.,Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jie Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China.,Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Chengbin Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Linqiang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Rendan Yang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Xue Jiang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yingjie Wu
- Shandong Provincial Hospital, Shandong Laboratory Animal Center, Shandong First Medical University and Shandong Academy of Medical Sciences. Jinan, Shandong, China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaoju Zou
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Bin Liang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| |
Collapse
|
26
|
Mekonnen D, Derbie A, Mihret A, Yimer SA, Tønjum T, Gelaw B, Nibret E, Munshae A, Waddell SJ, Aseffa A. Lipid droplets and the transcriptome of Mycobacterium tuberculosis from direct sputa: a literature review. Lipids Health Dis 2021; 20:129. [PMID: 34602073 PMCID: PMC8487580 DOI: 10.1186/s12944-021-01550-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/09/2021] [Indexed: 11/21/2022] Open
Abstract
Mycobacterium tuberculosis (Mtb), the main etiology of tuberculosis (TB), is predominantly an intracellular pathogen that has caused infection, disease and death in humans for centuries. Lipid droplets (LDs) are dynamic intracellular organelles that are found across the evolutionary tree of life. This review is an evaluation of the current state of knowledge regarding Mtb-LD formation and associated Mtb transcriptome directly from sputa.Based on the LD content, Mtb in sputum may be classified into three groups: LD positive, LD negative and LD borderline. However, the clinical and evolutionary importance of each state is not well elaborated. Mounting evidence supports the view that the presence of LD positive Mtb bacilli in sputum is a biomarker of slow growth, low energy state, towards lipid degradation, and drug tolerance. In Mtb, LD may serve as a source of chemical energy, scavenger of toxic compounds, prevent destruction of Mtb through autophagy, delay trafficking of lysosomes towards the phagosome, and contribute to Mtb persistence. It is suggest that LD is a key player in the induction of a spectrum of phenotypic and metabolic states of Mtb in the macrophage, granuloma and extracellular sputum microenvironment. Tuberculosis patients with high proportion of LD positive Mtb in pretreatment sputum was associated with higher rate of poor treatment outcome, indicating that LD may have a clinical application in predicting treatment outcome.The propensity for LD formation among Mtb lineages is largely unknown. The role of LD on Mtb transmission and disease phenotype (pulmonary TB vs extra-pulmonary TB) is not well understood. Thus, further studies are needed to understand the relationships between LD positivity and Mtb lineage, Mtb transmission and clinical types.
Collapse
Affiliation(s)
- Daniel Mekonnen
- Department of Medical Microbiology, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia.
- Institute of Biotechnology, Bahir Dar University, Bahir Dar, Ethiopia.
| | - Awoke Derbie
- Department of Medical Microbiology, College of Medicine and Health Sciences, Bahir Dar University, Bahir Dar, Ethiopia
- Institute of Biotechnology, Bahir Dar University, Bahir Dar, Ethiopia
- The Centre for Innovative Drug Development and Therapeutic Trials for Africa (CDT-Africa), Addis Ababa University, Addis Ababa, Ethiopia
| | - Adane Mihret
- Armauer Hansen Research Institute, Jimma Road, ALERT Compound, PO Box 1005, Addis Ababa, Ethiopia
- Department of Medical Microbiology, Immunology and Parasitology, College of Medicine and Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Solomon Abebe Yimer
- Department of Microbiology, University of Oslo, PO Box 1071, Blindern, NO-0316, Oslo, Norway
- Coalition for Epidemic Preparedness Innovations, CEPI, P.O. Box 123, Torshov, 0412, Oslo, Norway
| | - Tone Tønjum
- Department of Microbiology, University of Oslo, PO Box 1071, Blindern, NO-0316, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, PO Box 4950, Nydalen, NO-0424, Oslo, Norway
| | - Baye Gelaw
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Endalkachew Nibret
- Institute of Biotechnology, Bahir Dar University, Bahir Dar, Ethiopia
- Department of Biology, Bahir Dar University, Bahir Dar, Ethiopia
| | - Abaineh Munshae
- Institute of Biotechnology, Bahir Dar University, Bahir Dar, Ethiopia
- Department of Biology, Bahir Dar University, Bahir Dar, Ethiopia
| | - Simon J Waddell
- Department of Global Health and Infection, Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX, UK
| | - Abraham Aseffa
- Armauer Hansen Research Institute, Jimma Road, ALERT Compound, PO Box 1005, Addis Ababa, Ethiopia
| |
Collapse
|
27
|
Chen Q, Fang W, Cui K, Chen Q, Xiang X, Zhang J, Zhang Y, Mai K, Ai Q. Endoplasmic reticulum stress induces hepatic steatosis by transcriptional upregulating lipid droplet protein perilipin2. FASEB J 2021; 35:e21900. [PMID: 34547130 DOI: 10.1096/fj.202100739rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 08/12/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022]
Abstract
Previous studies have shown that endoplasmic reticulum (ER) stress contributes to hepatic steatosis in several manners. However, how lipid droplet (LD) proteins participate in this process has rarely been reported. In the present study, ER stress was induced at both in vitro and in vivo levels with tunicamycin in large yellow croaker (Larimichthys crocea). Effects of LD protein perilipin2 (PLIN2) on hepatic lipid accumulation and lipoprotein transport under normal physiological condition and ER stress were then explored using dsRNA mediated knockdown. Subsequently, the transcriptional regulation of plin2 expression by transcription factors generated in the unfolded protein response (UPR) was determined by dual-luciferase reporter assays, chromatin immunoprecipitation and electrophoretic mobility-shift assay. We demonstrated that ER stress could promote LDs accumulation and inhibit lipoprotein transport by transcriptionally upregulating PLIN2 in liver. Among the transcription factors generated by UPR, spliced X-box binding protein1 can directly upregulated the expression of plin2, whereas C/EBP homologous protein can upregulate the expression of plin2 through peroxisome proliferator activated-receptor α. These results revealed that the LD protein PLIN2 played an important role in ER stress-induced hepatic steatosis, which might be a novel mechanism explaining hepatic steatosis triggered by ER stress.
Collapse
Affiliation(s)
- Qiuchi Chen
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Wei Fang
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Kun Cui
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Qiang Chen
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Xiaojun Xiang
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Junzhi Zhang
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Yunqiang Zhang
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China
| | - Kangsen Mai
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, People's Republic of China
| | - Qinghui Ai
- Key laboratory of Aquaculture Nutrition and Feed, Ministry of Agriculture and Rural Affairs, and The Key Laboratory of Mariculture, Ministry of Education, Ocean University of China, Qingdao, People's Republic of China.,Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao, People's Republic of China
| |
Collapse
|
28
|
Caruso B, Wilke N, Perillo MA. Triglyceride Lenses at the Air-Water Interface as a Model System for Studying the Initial Stage in the Biogenesis of Lipid Droplets. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:10958-10970. [PMID: 34491757 DOI: 10.1021/acs.langmuir.1c01359] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Lipid droplets (LD) are intracellular structures consisting of an apolar lipid core, composed mainly of triglycerides (TG) and steryl esters, coated by a lipid-protein mixed monolayer. The mechanisms underlying LD biogenesis at the endoplasmic reticulum membrane are a matter of many current investigations. Although models explaining the budding-off of protuberances of phase-segregated TG inside bilayers have been proposed recently, the assumption of such initial blisters needs further empirical support. Here, we study mixtures of egg phosphatidylcholine (EPC) and TG at the air-water interface in order to describe some physical properties and topographic stability of TG bulk structures in contact with interfaces. Brewster angle microscopy images revealed the appearance of microscopic collapsed structures (CS) with highly reproducible lateral size (∼1 μm lateral radius) not varying with lateral packing changes and being highly stable at surface pressures (π) beyond collapse. By surface spectral fluorescence microscopy, we were able to characterize the solvatochromism of Nile Red both in monolayers and inside CS. This allowed to conclude that CS corresponded to a phase of liquid TG and to characterize them as lenses forming a three-phase (oil-water-air) system. Thereby, the thicknesses of the lenses could be determined, observing that they were dramatically flattened when EPC was present (6-12 nm compared to 30-50 nm for lenses on EPC/TG and TG films, respectively). Considering the shape of lenses, the interfacial tensions, and the Neumann's triangle, this experimental approach allows one to estimate the oil-water interfacial tension acting at each individual microscopic lens and at varying compression states of the surrounding monolayer. Thus, lenses formed on air-water Langmuir films can serve to assess variables of relevance to the initial step of LD biogenesis, such as the degree of dispersion of excluded-TG phase and shape, spatial distribution, and oil-water interfacial tension of lenses.
Collapse
Affiliation(s)
- B Caruso
- Facultad de Ciencias Exactas, Físicas y Naturales. Departamento de Química, Cátedra de Química BiológicaUniversidad Nacional de Córdoba, X5016GCA Córdoba, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET, X5016GCA Córdoba, Argentina
| | - N Wilke
- Facultad de Ciencias Químicas,. Departamento de Química Biológica Ranwel Caputto, Universidad Nacional de Córdoba, X5016GCA Córdoba, Argentina
- Centro de Investigaciones en Quimica Biológica de Córdoba (CIQUIBIC), CONICET, X5016GCA Córdoba, Argentina
| | - M A Perillo
- Facultad de Ciencias Exactas, Físicas y Naturales. Departamento de Química, Cátedra de Química BiológicaUniversidad Nacional de Córdoba, X5016GCA Córdoba, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET, X5016GCA Córdoba, Argentina
| |
Collapse
|
29
|
Ma X, Zhi Z, Zhang S, Zhou C, Mechler A, Liu P. Validating an artificial organelle: Studies of lipid droplet-specific proteins on adiposome platform. iScience 2021; 24:102834. [PMID: 34368652 PMCID: PMC8326204 DOI: 10.1016/j.isci.2021.102834] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/28/2021] [Accepted: 07/08/2021] [Indexed: 10/25/2022] Open
Abstract
New strategies are urgently needed to characterize the functions of the lipid droplet (LD). Here, adiposome, an artificial LD mimetic platform, was validated by comparative in vitro bioassays. Scatchard analysis found that the binding of perilipin 2 (PLIN2) to the adiposome surface was saturable. Phosphatidylinositol (PtdIns) was found to inhibit PLIN2 binding while it did not impede perilipin 3 (PLIN3). Structural analysis combined with mutagenesis revealed that the 73rd glutamic acid of PLIN2 is significant for the effect of PtdIns on the PLIN2 binding. Furthermore, adiposome was also found to be an ideal platform for in situ enzymatic activity measurement of adipose triglyceride lipase (ATGL). The significant serine mutants of ATGL were found to cause the loss of lipase activity. Our study demonstrates the adiposome as a powerful, manipulatable model system that mimics the function of LD for binding and enzymatic activity studies of LD proteins in vitro.
Collapse
Affiliation(s)
- Xuejing Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zelun Zhi
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, Australia
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chang Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Adam Mechler
- Department of Chemistry and Physics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, 3086, Australia
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
30
|
Biophenolic Profile Modulations in Olive Tissues as Affected by Manganese Nutrition. PLANTS 2021; 10:plants10081724. [PMID: 34451769 PMCID: PMC8402200 DOI: 10.3390/plants10081724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/10/2021] [Accepted: 08/16/2021] [Indexed: 11/23/2022]
Abstract
Manganese (Mn) is an essential element that intervenes in several plant metabolic processes. The olive tree, and its fruits and leaves, are known as a source of nutraceuticals since they are rich in biophenols. However, there is still a serious lack of data about biophenolic distribution in olive stems and roots under Mn fertilisation. In this context, our study aimed to examine the effects of Mn fertilisation on the biophenolic profile in the leaves, stems, and roots of the ‘Istarska bjelica’ olive cultivar. The experiment was set up in a greenhouse, during a period of five months, as a random block design consisting of three treatments with varying Mn concentrations in full-strength Hoagland’s nutrient solution (0.2 µM Mn, 12 µM Mn, and 24 µM Mn). The obtained results indicate that the amount of Mn in the examined olive plant tissues was significantly higher under 12 µM Mn and 24 µM Mn treatments compared to that of the 0.2 µM Mn treatment. While the concentration of biophenols varied in roots depending on the compound in question, a strong positive impact of the increased Mn concentration in nutrient solution (12 µM Mn and 24 µM Mn) on the concentrations of the main biophenolic compounds was observed in stems. The concentration of oleuropein in leaves almost doubled at 24 µM Mn, with the highest Mn concentration, as compared to the 0.2 µM Mn treatment. The obtained results led to the conclusion that the supply of Mn could enhance the concentration of some biologically active compounds in olives grown hydroponically, implying a critical need for further investigation of Mn fertilisation practices in the conventional olive farming system.
Collapse
|
31
|
Bosch M, Sweet MJ, Parton RG, Pol A. Lipid droplets and the host-pathogen dynamic: FATal attraction? J Cell Biol 2021; 220:e202104005. [PMID: 34165498 PMCID: PMC8240858 DOI: 10.1083/jcb.202104005] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 02/06/2023] Open
Abstract
In the ongoing conflict between eukaryotic cells and pathogens, lipid droplets (LDs) emerge as a choke point in the battle for nutrients. While many pathogens seek the lipids stored in LDs to fuel an expensive lifestyle, innate immunity rewires lipid metabolism and weaponizes LDs to defend cells and animals. Viruses, bacteria, and parasites directly and remotely manipulate LDs to obtain substrates for metabolic energy, replication compartments, assembly platforms, membrane blocks, and tools for host colonization and/or evasion such as anti-inflammatory mediators, lipoviroparticles, and even exosomes. Host LDs counterattack such advances by synthesizing bioactive lipids and toxic nucleotides, organizing immune signaling platforms, and recruiting a plethora of antimicrobial proteins to provide a front-line defense against the invader. Here, we review the current state of this conflict. We will discuss why, when, and how LDs efficiently coordinate and precisely execute a plethora of immune defenses. In the age of antimicrobial resistance and viral pandemics, understanding innate immune strategies developed by eukaryotic cells to fight and defeat dangerous microorganisms may inform future anti-infective strategies.
Collapse
Affiliation(s)
- Marta Bosch
- Lipid Trafficking and Disease Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Matthew J. Sweet
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, Australia
| | - Albert Pol
- Lipid Trafficking and Disease Group, Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| |
Collapse
|
32
|
Brink JTR, Fourie R, Sebolai O, Albertyn J, Pohl CH. The role of lipid droplets in microbial pathogenesis. J Med Microbiol 2021; 70. [PMID: 34184983 DOI: 10.1099/jmm.0.001383] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The nonpolar lipids present in cells are mainly triacylglycerols and steryl esters. When cells are provided with an abundance of nutrients, these storage lipids accumulate. As large quantities of nonpolar lipids cannot be integrated into membranes, they are isolated from the cytosolic environment in lipid droplets. As specialized, inducible cytoplasmic organelles, lipid droplets have functions beyond the regulation of lipid metabolism, in cell signalling and activation, membrane trafficking and control of inflammatory mediator synthesis and secretion. Pathogens, including fungi, viruses, parasites, or intracellular bacteria can induce and may benefit from lipid droplets in infected cells. Here we review biogenesis of lipid droplets as well as the role of lipid droplets in the pathogenesis of selected viruses, bacteria, protists and yeasts.
Collapse
Affiliation(s)
- Jacobus T R Brink
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Ruan Fourie
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Olihile Sebolai
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Jacobus Albertyn
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| | - Carolina H Pohl
- Department of Microbiology and Biochemistry, University of the Free State, Bloemfontein, South Africa
| |
Collapse
|
33
|
Deng Y, Zhou C, Mirza AH, Bamigbade AT, Zhang S, Xu S, Liu P. Rab18 binds PLIN2 and ACSL3 to mediate lipid droplet dynamics. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158923. [PMID: 33713834 DOI: 10.1016/j.bbalip.2021.158923] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 02/26/2021] [Accepted: 03/05/2021] [Indexed: 01/16/2023]
Abstract
Lipid droplet (LD) is a vital organelle governing lipid homeostasis and Rab18 has been linked to lipid metabolism. However, the mechanisms of Rab18-mediated LD dynamics in myoblast cells remain elusive. Here, we report that Rab18 plays an important role in oleic acid (OA)-induced LD accumulation in mouse myoblast C2C12 cells. Rab18 was translocated from the endoplasmic reticulum (ER) to LDs during LD accumulation, which was regulated by perilipin 2 (PLIN2), a major LD protein. LD-associated Rab18 bound with the C terminus of PLIN2 and the LD localization of Rab18 was diminished when PLIN2 was depleted. Moreover, loss of function of Rab18 led to reduced triacylglycerol (TAG) level and fewer but larger LDs. In contrast, overexpression of Rab18 resulted in elevated TAG content and LD number. Furthermore, LD-associated Rab18 interacted with acyl-CoA synthetase long-chain family member 3 (ACSL3), which in turn promoted the LD localization of this protein. These data show that Rab18 interacts with PLIN2 and forms a complex with PLIN2 and ACSL3, which plays a critical role in LD accumulation and dynamics of myoblast cells.
Collapse
Affiliation(s)
- Yaqin Deng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chang Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Ahmed Hammad Mirza
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Adekunle T Bamigbade
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
34
|
Feringa FM, van der Kant R. Cholesterol and Alzheimer's Disease; From Risk Genes to Pathological Effects. Front Aging Neurosci 2021; 13:690372. [PMID: 34248607 PMCID: PMC8264368 DOI: 10.3389/fnagi.2021.690372] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/28/2021] [Indexed: 12/22/2022] Open
Abstract
While the central nervous system compromises 2% of our body weight, it harbors up to 25% of the body's cholesterol. Cholesterol levels in the brain are tightly regulated for physiological brain function, but mounting evidence indicates that excessive cholesterol accumulates in Alzheimer's disease (AD), where it may drive AD-associated pathological changes. This seems especially relevant for late-onset AD, as several of the major genetic risk factors are functionally associated with cholesterol metabolism. In this review we discuss the different systems that maintain brain cholesterol metabolism in the healthy brain, and how dysregulation of these processes can lead, or contribute to, Alzheimer's disease. We will also discuss how AD-risk genes might impact cholesterol metabolism and downstream AD pathology. Finally, we will address the major outstanding questions in the field and how recent technical advances in CRISPR/Cas9-gene editing and induced pluripotent stem cell (iPSC)-technology can aid to study these problems.
Collapse
Affiliation(s)
- Femke M. Feringa
- Department of Clinical Genetics, Center for Neurogenomics and Cognitive Research (CNCR), Amsterdam University Medical Center, Amsterdam, Netherlands
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, Netherlands
| | - Rik van der Kant
- Department of Functional Genomics, Center for Neurogenomics and Cognitive Research (CNCR), VU University Amsterdam, Amsterdam, Netherlands
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
35
|
Mantilla MJ, Cabrera Díaz CE, Ariza-Aranguren G, de Cock H, Helms JB, Restrepo S, Jiménez E, Celis Ramírez AM. Back to the Basics: Two Approaches for the Identification and Extraction of Lipid Droplets from Malassezia pachydermatis CBS1879 and Malassezia globosa CBS7966. Curr Protoc 2021; 1:e122. [PMID: 33950584 DOI: 10.1002/cpz1.122] [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] [Indexed: 12/21/2022]
Abstract
Malassezia spp. are lipid-dependent yeasts that have been related to skin mycobiota and dermatological and systemic diseases. Study of lipid droplets (LDs) is relevant to elucidate the unknown role of these organelles in Malassezia and to gain a broader overview of lipid metabolism in Malassezia. Here, we standardized two protocols for the analysis of LDs in M. pachydermatis and M. globosa. The first describes co-staining for confocal laser-scanning fluorescence microscopy, and the second details extraction and purification of LDs. The double stain is achieved with three different neutral lipid fluorophores, namely Nile Red, BODIPY™ 493/503, and HCS LipidTOX™ Deep Red Neutral, in combination with Calcofluor White. For LD extraction, cell wall rupture is conducted using Trichoderma harzianum enzymes and cycles of vortexing with zirconium beads. LD purification is performed in a three-step ultracentrifugation process. These standardizations will contribute to the study of the dynamics, morphology, and composition of LDs in Malassezia. © 2021 Wiley Periodicals LLC. Basic Protocol 1: Lipid droplet fluorescence staining Basic Protocol 2: Lipid droplet extraction and purification Support Protocol: Malassezia spp. culture conditions.
Collapse
Affiliation(s)
- Maria Juliana Mantilla
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Department of Biological Sciences, Universidad de Los Andes, Bogotá, Colombia
| | | | - Gabriela Ariza-Aranguren
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Department of Biological Sciences, Universidad de Los Andes, Bogotá, Colombia
| | - Hans de Cock
- Microbiology, Department of Biology, Faculty of Science, Institute of Biomembranes, Utrecht University, Utrecht, The Netherlands
| | - J Bernd Helms
- Department of Biomolecular Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - Silvia Restrepo
- Laboratorio de Micología y Fitopatología (LAMFU), Chemical and Food Engineering Department, Universidad de Los Andes, Bogotá, Colombia
| | - Elizabeth Jiménez
- Applied Biochemistry Research Group (GIBA), Department of Chemistry, Universidad de Los Andes, Bogotá, Colombia
| | - Adriana Marcela Celis Ramírez
- Grupo de Investigación Celular y Molecular de Microorganismos Patógenos (CeMoP), Department of Biological Sciences, Universidad de Los Andes, Bogotá, Colombia
| |
Collapse
|
36
|
Bart G, Fischer D, Samoylenko A, Zhyvolozhnyi A, Stehantsev P, Miinalainen I, Kaakinen M, Nurmi T, Singh P, Kosamo S, Rannaste L, Viitala S, Hiltunen J, Vainio SJ. Characterization of nucleic acids from extracellular vesicle-enriched human sweat. BMC Genomics 2021; 22:425. [PMID: 34103018 PMCID: PMC8188706 DOI: 10.1186/s12864-021-07733-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/17/2021] [Indexed: 01/08/2023] Open
Abstract
Background The human sweat is a mixture of secretions from three types of glands: eccrine, apocrine, and sebaceous. Eccrine glands open directly on the skin surface and produce high amounts of water-based fluid in response to heat, emotion, and physical activity, whereas the other glands produce oily fluids and waxy sebum. While most body fluids have been shown to contain nucleic acids, both as ribonucleoprotein complexes and associated with extracellular vesicles (EVs), these have not been investigated in sweat. In this study we aimed to explore and characterize the nucleic acids associated with sweat particles. Results We used next generation sequencing (NGS) to characterize DNA and RNA in pooled and individual samples of EV-enriched sweat collected from volunteers performing rigorous exercise. In all sequenced samples, we identified DNA originating from all human chromosomes, but only the mitochondrial chromosome was highly represented with 100% coverage. Most of the DNA mapped to unannotated regions of the human genome with some regions highly represented in all samples. Approximately 5 % of the reads were found to map to other genomes: including bacteria (83%), archaea (3%), and virus (13%), identified bacteria species were consistent with those commonly colonizing the human upper body and arm skin. Small RNA-seq from EV-enriched pooled sweat RNA resulted in 74% of the trimmed reads mapped to the human genome, with 29% corresponding to unannotated regions. Over 70% of the RNA reads mapping to an annotated region were tRNA, while misc. RNA (18,5%), protein coding RNA (5%) and miRNA (1,85%) were much less represented. RNA-seq from individually processed EV-enriched sweat collection generally resulted in fewer percentage of reads mapping to the human genome (7–45%), with 50–60% of those reads mapping to unannotated region of the genome and 30–55% being tRNAs, and lower percentage of reads being rRNA, LincRNA, misc. RNA, and protein coding RNA. Conclusions Our data demonstrates that sweat, as all other body fluids, contains a wealth of nucleic acids, including DNA and RNA of human and microbial origin, opening a possibility to investigate sweat as a source for biomarkers for specific health parameters. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-07733-9.
Collapse
Affiliation(s)
- Geneviève Bart
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Daniel Fischer
- Production Systems, Natural Resources Institute Finland (LUKE), 31600, Jokioinen, Finland
| | - Anatoliy Samoylenko
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Artem Zhyvolozhnyi
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Pavlo Stehantsev
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Ilkka Miinalainen
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Mika Kaakinen
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Tuomas Nurmi
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Prateek Singh
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland.,Present Address: Finnadvance, Aapistie 5, 90220, Oulu, Finland
| | - Susanna Kosamo
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland
| | - Lauri Rannaste
- Biosensors, VTT, Technical Research Center of Finland Ltd, Kaitoväylä 1, 90570, Oulu, Finland
| | - Sirja Viitala
- Production Systems, Natural Resources Institute Finland (LUKE), 31600, Jokioinen, Finland
| | - Jussi Hiltunen
- Biosensors, VTT, Technical Research Center of Finland Ltd, Kaitoväylä 1, 90570, Oulu, Finland
| | - Seppo J Vainio
- Faculty of Biochemistry and Molecular Medicine, Disease Networks Research Unit, Laboratory of Developmental Biology, Kvantum Institute, Infotech Oulu, University of Oulu, 90014 University of Oulu, Oulu, Finland.
| |
Collapse
|
37
|
Tegeder I, Kögel D. When lipid homeostasis runs havoc: Lipotoxicity links lysosomal dysfunction to autophagy. Matrix Biol 2021; 100-101:99-117. [DOI: 10.1016/j.matbio.2020.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/29/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
|
38
|
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: 1.7] [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
|
39
|
Yan M, Wu Y, Peng W, Fu C, Giunta S, Chang M, Zhang G, Dou M, Xia S, Li H, Zhou J, Shen Y. Exposure to particulate matter 2.5 and cigarette smoke induces the synthesis of lipid droplets by glycerol kinase 5. Clin Exp Pharmacol Physiol 2021; 48:498-507. [PMID: 33462866 DOI: 10.1111/1440-1681.13463] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/26/2020] [Indexed: 12/20/2022]
Abstract
Particulate matter (PM2.5) and cigarette smoke exposure are leading factors contributing to various diseases, especially respiratory diseases. The purpose of this research was to study the effects of PM2.5 and cigarette smoke on glycerol kinase 5 (GK5) expression and the possible mechanisms by which GK5 participates in lipid droplet (LD) synthesis in alveolar epithelial A549 cells. Real-time polymerase chain reaction (RT-PCR) and western blotting have been used for the detection of messenger RNA (mRNA) and protein expression respectively. GK5 overexpressing cells were established by lentivirus transfection, whereby lentiviral vectors deliver the gene into chromosomes, allowing stable expression. Affymetrix microarray analysis, a widely used tool for measuring genome-wide gene expression, has been used to explore differential gene expression profiles. A549 cells stimulated with PM2.5 and cigarette smoke extract (CSE) showed elevated GK5 expression in a dose-dependent manner. Transmission electron microscopy and oil red O staining were used to observe LDs in cells. Further, GK5 overexpressing cells showed increased LDs and upregulation of genes and proteins related to lipogenesis and lipid transportation. Affymetrix microarray analysis revealed that GK5 overexpression resulted in the differential expression of more than 109 genes, which were mainly involved in the regulation of cell death, cell survival, cellular movement and migration, and those involved in cellular growth and proliferation pathways. Overall, this study demonstrates that GK5 is upregulated during PM2.5 and cigarette smoke exposure and induces LD synthesis.
Collapse
Affiliation(s)
- Mengnan Yan
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuanyuan Wu
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenjun Peng
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cuiping Fu
- Department of Pulmonary Medicine, The First Hospital Affiliated to Soochow University, Suzhou, China
| | - Sergio Giunta
- Casa di Cura Prof. Nobili-GHC Garofalo Health Care, Bologna, Italy
| | - Meijia Chang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ge Zhang
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Maosen Dou
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shijin Xia
- Department of Geriatrics, Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, China
| | - Huayin Li
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Pulmonary Medicine, Shanghai Respiratory Research Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yao Shen
- Department of Pulmonary Medicine, Pudong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
40
|
Huang T, Bamigbade AT, Xu S, Deng Y, Xie K, Ogunsade OO, Mirza AH, Wang J, Liu P, Zhang S. Identification of noncoding RNA-encoded proteins on lipid droplets. Sci Bull (Beijing) 2021; 66:314-318. [PMID: 36654408 DOI: 10.1016/j.scib.2020.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 01/20/2023]
Affiliation(s)
- Ting Huang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Adekunle T Bamigbade
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yaqin Deng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kang Xie
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ololade O Ogunsade
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ahmed Hammad Mirza
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jifeng Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Pingsheng Liu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Shuyan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
41
|
Rogers S, Henne WM. Analysis of Neutral Lipid Synthesis in Saccharomyces cerevisiae by Metabolic Labeling and Thin Layer Chromatography. J Vis Exp 2021. [PMID: 33616103 DOI: 10.3791/62201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Neutral lipids (NLs) are a class of hydrophobic, chargeless biomolecules that play key roles in energy and lipid homeostasis. NLs are synthesized de novo from acetyl-CoA and are primarily present in eukaryotes in the form of triglycerides (TGs) and sterol-esters (SEs). The enzymes responsible for the synthesis of NLs are highly conserved from Saccharomyces cerevisiae (yeast) to humans, making yeast a useful model organism to dissect the function and regulation of NL metabolism enzymes. While much is known about how acetyl-CoA is converted into a diverse set of NL species, mechanisms for regulating NL metabolism enzymes, and how mis-regulation can contribute to cellular pathologies, are still being discovered. Numerous methods for the isolation and characterization of NL species have been developed and used over decades of research; however, a quantitative and simple protocol for the comprehensive characterization of major NL species has not been discussed. Here, a simple and adaptable method to quantify the de novo synthesis of major NL species in yeast is presented. We apply 14C-acetic acid metabolic labeling coupled with thin layer chromatography to separate and quantify a diverse range of physiologically important NLs. Additionally, this method can be easily applied to study in vivo reaction rates of NL enzymes or degradation of NL species over time.
Collapse
Affiliation(s)
- Sean Rogers
- Department of Cell Biology, University of Texas Southwestern Medical Center
| | - W Mike Henne
- Department of Cell Biology, University of Texas Southwestern Medical Center;
| |
Collapse
|
42
|
Cloherty AP, Olmstead AD, Ribeiro CM, Jean F. Hijacking of Lipid Droplets by Hepatitis C, Dengue and Zika Viruses-From Viral Protein Moonlighting to Extracellular Release. Int J Mol Sci 2020; 21:E7901. [PMID: 33114346 PMCID: PMC7662613 DOI: 10.3390/ijms21217901] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 02/06/2023] Open
Abstract
Hijacking and manipulation of host cell biosynthetic pathways by human enveloped viruses are essential for the viral lifecycle. Flaviviridae members, including hepatitis C, dengue and Zika viruses, extensively manipulate host lipid metabolism, underlining the importance of lipid droplets (LDs) in viral infection. LDs are dynamic cytoplasmic organelles that can act as sequestration platforms for a unique subset of host and viral proteins. Transient recruitment and mobilization of proteins to LDs during viral infection impacts host-cell biological properties, LD functionality and canonical protein functions. Notably, recent studies identified LDs in the nucleus and also identified that LDs are transported extracellularly via an autophagy-mediated mechanism, indicating a novel role for autophagy in Flaviviridae infections. These developments underline an unsuspected diversity and localization of LDs and potential moonlighting functions of LD-associated proteins during infection. This review summarizes recent breakthroughs concerning the LD hijacking activities of hepatitis C, dengue and Zika viruses and potential roles of cytoplasmic, nuclear and extracellular LD-associated viral proteins during infection.
Collapse
Affiliation(s)
- Alexandra P.M. Cloherty
- Amsterdam UMC, Amsterdam Institute for Infection & Immunity, Department of Experimental Immunology, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.P.M.C.); (C.M.S.R.)
| | - Andrea D. Olmstead
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, 3559–2350 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada;
| | - Carla M.S. Ribeiro
- Amsterdam UMC, Amsterdam Institute for Infection & Immunity, Department of Experimental Immunology, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (A.P.M.C.); (C.M.S.R.)
| | - François Jean
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, 3559–2350 Health Sciences Mall, Vancouver, BC V6T1Z3, Canada;
| |
Collapse
|
43
|
Seena S, Sobral O, Cano A. Metabolomic, functional, and ecologic responses of the common freshwater fungus Neonectria lugdunensis to mine drainage stress. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 718:137359. [PMID: 32092520 DOI: 10.1016/j.scitotenv.2020.137359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/14/2020] [Accepted: 02/14/2020] [Indexed: 06/10/2023]
Abstract
Metal contamination of watersheds is a global problem. Here, we conducted litter decomposition studies with Neonectria lugdunensis, a cosmopolitan aquatic fungus. Fungal isolates from four reference (non-impacted) and six metal-contaminated streams (due to mine drainage) were exposed to mine drainage and reference stream waters in Central Portugal. Impact of mine drainage waters on N. lugdunensis hyphae was investigated by performing metabolomic profiling of 200 lipids and 25 amino acids (AA) with ultra-high performance liquid chromatography-mass spectrometry. In parallel, functional response of N. lugdunensis isolates was assessed through expression profiles of a functional gene, cellobiohydrolase I (CbhI). Ecological performance via leaf mass loss was also determined. Exposure to mine drainage waters altered the concentration of numerous AA and lipids. Most strikingly, a gradual increase in the concentration of the triacylglycerols (TAG) with shorter acyl chains and lesser unsaturation was observed after the exposure to mine drainage waters. In addition, the changes in the concentration of numerous TAG, lysophosphatidylcholines, and AA were more significant in the isolates from the metal-contaminated streams after exposure to mine drainage water. CbhI gene of the isolates from reference streams was down-regulated by metal stress, while those from metal-contaminated streams remained unaffected. Finally, leaf mass loss was influenced by both exposure to mine drainage waters and the origin of isolates. Overall, our study demonstrates unique functional signatures displayed by fungi under metal stress and the relevant role that fungal AA and lipids play to cope with metal toxicity.
Collapse
Affiliation(s)
- Sahadevan Seena
- MARE - Marine and Environmental Sciences Centre, University of Coimbra, PT-3004-517 Coimbra, Portugal; CBMA - Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal.
| | - Olímpia Sobral
- MARE - Marine and Environmental Sciences Centre, University of Coimbra, PT-3004-517 Coimbra, Portugal; CBMA - Centre of Molecular and Environmental Biology, Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Ainara Cano
- ONE WAY LIVER, S.L., Parque Tecnológico de Bizkaia, edif.502- plta 0, 48160 Derio, Bizkaia, Spain
| |
Collapse
|
44
|
Kubota N, Ojima H, Hatano M, Yamazaki K, Masugi Y, Tsujikawa H, Fujii-Nishimura Y, Ueno A, Kurebayashi Y, Shinoda M, Kitago M, Abe Y, Kitagawa Y, Sakamoto M. Clinicopathological features of hepatocellular carcinoma with fatty change: Tumors with macrovesicular steatosis have better prognosis and aberrant expression patterns of perilipin and adipophilin. Pathol Int 2020; 70:199-209. [PMID: 31930673 DOI: 10.1111/pin.12889] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 11/26/2019] [Indexed: 12/19/2022]
Abstract
The clinicopathological characteristics of steatosis in hepatocellular carcinoma (HCC) remain unclear. Here, we elucidate the features of macrovesicular steatosis (MaS) and microvesicular steatosis (MiS) in HCC and their relationships with background liver steatosis. A total of 165 HCC lesions were classified as MaS-HCC, MiS-HCC, or conventional HCC (cHCC) according to the cutoff value of 30% MaS or MiS in tumor cells. We analyzed the clinicopathological differences among these groups. MaS-HCC had less portal vein invasion, a higher proportion of HCC with intratumoral fibrosis, and a lower cumulative risk of recurrence than MiS-HCC or cHCC. Moreover, both MaS-HCC and MiS-HCC had lower incidences of hepatitis virus infection and higher levels of HbA1c than cHCC. Background liver steatosis was also higher in MaS-HCC than in cHCC. Immunohistochemical expression of perilipin (Plin1) and adipophilin (ADRP), major proteins expressed on lipid droplet membranes, revealed that almost all lipid droplets in HCC were Plin1 negative, whereas those in background liver were positive. In contrast, ADRP was expressed on lipid droplets in both HCC and background liver. We concluded that MaS-HCC and MiS-HCC were associated with metabolic abnormalities but exhibited different biologic behaviors. Furthermore, lipid droplets in HCC were pathophysiologically different from those in background liver.
Collapse
Affiliation(s)
- Naoto Kubota
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Ojima
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Mami Hatano
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.,Department of Pathology, National Hospital Organization, Tokyo Medical Center, Tokyo, Japan
| | - Ken Yamazaki
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yohei Masugi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Hanako Tsujikawa
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | | | - Akihisa Ueno
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kurebayashi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Masahiro Shinoda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuta Abe
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Michiie Sakamoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
45
|
Steryl Ester Formation and Accumulation in Steroid-Degrading Bacteria. Appl Environ Microbiol 2020; 86:AEM.02353-19. [PMID: 31704679 DOI: 10.1128/aem.02353-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 11/01/2019] [Indexed: 11/20/2022] Open
Abstract
Steryl esters (SEs) are important storage compounds in many eukaryotes and are often prominent components of intracellular lipid droplets. Here, we demonstrate that selected Actino- and Proteobacteria growing on sterols are also able to synthesize SEs and to sequester them in cytoplasmic lipid droplets. We found cholesteryl ester (CE) formation in members of the actinobacterial genera Rhodococcus, Mycobacterium, and Amycolatopsis, as well as several members of the proteobacterial Cellvibrionales order. CEs maximally accumulated under nitrogen-limiting conditions, suggesting that steryl ester formation plays a crucial role for storing excess energy and carbon under adverse conditions. Rhodococcus jostii RHA1 was able to synthesize phytosteryl and cholesteryl esters, the latter reaching up to 7% of its cellular dry weight and 69% of its lipid droplets. Purified lipid droplets from RHA1 contained CEs, free cholesterol, and triacylglycerols. In addition, we found formation of CEs in Mycobacterium tuberculosis when it was grown with cholesterol plus an additional fatty acid substrate. This study provides a basis for the application of bacterial whole-cell systems in the biotechnological production of SEs for use in functional foods and cosmetics.IMPORTANCE Oleaginous bacteria exhibit great potential for the production of high-value neutral lipids, such as triacylglycerols and wax esters. This study describes the formation of steryl esters (SEs) as neutral lipid storage compounds in sterol-degrading oleaginous bacteria, providing a basis for biotechnological production of SEs using bacterial systems with potential applications in the functional food, nutraceutical, and cosmetic industries. We found cholesteryl ester (CE) formation in several sterol-degrading Actino- and Proteobacteria under nitrogen-limiting conditions, suggesting an important role of this process in storing energy and carbon under adverse conditions. In addition, Mycobacterium tuberculosis grown on cholesterol accumulated CEs in the presence of an additional fatty acid substrate.
Collapse
|
46
|
The Puzzling Conservation and Diversification of Lipid Droplets from Bacteria to Eukaryotes. Results Probl Cell Differ 2020; 69:281-334. [PMID: 33263877 DOI: 10.1007/978-3-030-51849-3_11] [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] [Indexed: 01/17/2023]
Abstract
Membrane compartments are amongst the most fascinating markers of cell evolution from prokaryotes to eukaryotes, some being conserved and the others having emerged via a series of primary and secondary endosymbiosis events. Membrane compartments comprise the system limiting cells (one or two membranes in bacteria, a unique plasma membrane in eukaryotes) and a variety of internal vesicular, subspherical, tubular, or reticulated organelles. In eukaryotes, the internal membranes comprise on the one hand the general endomembrane system, a dynamic network including organelles like the endoplasmic reticulum, the Golgi apparatus, the nuclear envelope, etc. and also the plasma membrane, which are linked via direct lateral connectivity (e.g. between the endoplasmic reticulum and the nuclear outer envelope membrane) or indirectly via vesicular trafficking. On the other hand, semi-autonomous organelles, i.e. mitochondria and chloroplasts, are disconnected from the endomembrane system and request vertical transmission following cell division. Membranes are organized as lipid bilayers in which proteins are embedded. The budding of some of these membranes, leading to the formation of the so-called lipid droplets (LDs) loaded with hydrophobic molecules, most notably triacylglycerol, is conserved in all clades. The evolution of eukaryotes is marked by the acquisition of mitochondria and simple plastids from Gram-positive bacteria by primary endosymbiosis events and the emergence of extremely complex plastids, collectively called secondary plastids, bounded by three to four membranes, following multiple and independent secondary endosymbiosis events. There is currently no consensus view of the evolution of LDs in the Tree of Life. Some features are conserved; others show a striking level of diversification. Here, we summarize the current knowledge on the architecture, dynamics, and multitude of functions of the lipid droplets in prokaryotes and in eukaryotes deriving from primary and secondary endosymbiosis events.
Collapse
|
47
|
Zhu Y, Ren C, Zhang M, Zhong Y. Perilipin 5 Reduces Oxidative Damage Associated With Lipotoxicity by Activating the PI3K/ERK-Mediated Nrf2-ARE Signaling Pathway in INS-1 Pancreatic β-Cells. Front Endocrinol (Lausanne) 2020; 11:166. [PMID: 32296390 PMCID: PMC7136399 DOI: 10.3389/fendo.2020.00166] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 03/09/2020] [Indexed: 12/16/2022] Open
Abstract
Oxidative stress induced by free fatty acid overload in pancreatic β-cells is a potential contributory factor to dysfunction of insulin secretion and apoptotic cell death. Perilipin 5 (Plin5) has been reported to ameliorate oxidative stress-mediated damage in non-insulin-secreting tissues. We tested the hypothesis that Plin5 plays a similar role in pancreatic β-cells, which are extremely sensitive to oxidative stress. Here, our in vitro data showed that Plin5-mediated alleviation of palmitate-triggered apoptosis involves the mitochondrial pathway. And the protective role of Plin5 on β-cells was partially dependent on its modulation in oxidative stress. Upregulation of Plin5 in INS-1 cells decreased reactive oxygen species production, enhanced cellular glutathione levels, and induced expression of antioxidant enzymes glutamate-cysteine ligase catalytic subunit and heme oxygenase-1. However, knocking out of Plin5 abolished all of these beneficial effects. Furthermore, by using the O2- scavenger MnTMPyP, we verified that altering Plin5 expression impacted lipotoxic cell death partially via modulating oxidative stress. Mechanistic experiments revealed that Plin5 induced Nrf2-ARE system, a master regulator in the cellular adaptive response to oxidative stress, by activating PI3K/Akt and ERK signal pathways, contributing to the increase of antioxidant defense and consequently improving β-cell function and survival in the presence of lipotoxic oxidative stress. Overall, our findings indicate that Plin5 abrogates oxidative damage in INS-1 β-cells during lipotoxic stress partially through the enhancement of antioxidant defense involving the PI3K/Akt and ERK mediated Nrf2-ARE system.
Collapse
Affiliation(s)
- Yunxia Zhu
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
- *Correspondence: Yunxia Zhu
| | - Chenxi Ren
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Mingliang Zhang
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuan Zhong
- Department of Geriatrics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
48
|
Tan Y, Jin Y, Wu X, Ren Z. PSMD1 and PSMD2 regulate HepG2 cell proliferation and apoptosis via modulating cellular lipid droplet metabolism. BMC Mol Biol 2019; 20:24. [PMID: 31703613 PMCID: PMC6842266 DOI: 10.1186/s12867-019-0141-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 10/29/2019] [Indexed: 01/18/2023] Open
Abstract
Background Obesity and nonalcoholic steatohepatitis (NASH) are well-known risk factors of hepatocellular carcinoma (HCC). The lipid-rich environment enhances the proliferation and metastasis abilities of tumor cells. Previous studies showed the effect of the ubiquitin–proteasome system (UPS) on tumor cell proliferation. However, the underlying mechanism of UPS in regulating the proliferation of lipid-rich tumor cells is not totally clear. Results Here, we identify two proteasome 26S subunits, non-ATPase 1 and 2 (PSMD1 and PSMD2), which regulate HepG2 cells proliferation via modulating cellular lipid metabolism. Briefly, the knockdown of PSMD1 and/or PSMD2 decreases the formation of cellular lipid droplets, the provider of the energy and membrane components for tumor cell proliferation. Mechanically, PSMD1 and PSMD2 regulate the expression of genes related to de novo lipid synthesis via p38-JNK and AKT signaling. Moreover, the high expression of PSMD1 and PSMD2 is significantly correlated with poor prognosis of HCC. Conclusion We demonstrate that PSMD1 and PSMD2 promote the proliferation of HepG2 cells via facilitating cellular lipid droplet accumulation. This study provides a potential therapeutic strategy for the treatment of lipid-rich tumors.
Collapse
Affiliation(s)
- Yanjie Tan
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Yi Jin
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Xiang Wu
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China
| | - Zhuqing Ren
- Key Laboratory of Swine Genetics and Breeding of Ministry of Agriculture & Key Laboratory of Agriculture Animal Genetics, Breeding and Reproduction of Ministry of Education, College of Animal Science, Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China. .,Bio-Medical Center of Huazhong Agricultural University, Wuhan, 430070, Hubei, People's Republic of China.
| |
Collapse
|
49
|
Yu J, Zhang L, Li Y, Zhu X, Xu S, Zhou XM, Wang H, Zhang H, Liang B, Liu P. The Adrenal Lipid Droplet is a New Site for Steroid Hormone Metabolism. Proteomics 2019; 18:e1800136. [PMID: 30358111 DOI: 10.1002/pmic.201800136] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 10/08/2018] [Indexed: 01/25/2023]
Abstract
Steroid hormones play essential roles for living organisms. It has been long and well established that the endoplasmic reticulum (ER) and mitochondria are essential sites for steroid hormone biosynthesis because several steroidogenic enzymes are located in these organelles. The adrenal gland lipid droplet (LD) proteomes from human, macaque monkey, and rodent are analyzed, revealing that steroidogenic enzymes are also present in abundance on LDs. The enzymes found include 3β-hydroxysteroid dehydrogenase (HSD3B) and estradiol 17β-dehydrogenase 11 (HSD17B11). Analyses by Western blot and subcellular localization consistently demonstrate that HSD3B2 is localized on LDs. Furthermore, in vitro experiments confirm that the isolated LDs from HeLa cell stably expressing HSD3B2 or from rat adrenal glands have the capacity to convert pregnenolone to progesterone. Collectively, these data suggest that LDs may be important sites of steroid hormone metabolism. These findings may bring novel insights into the biosynthesis and metabolism of steroid hormones and the development of treatments for adrenal disorders.
Collapse
Affiliation(s)
- Jinhai Yu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Institute of Biophysics, Beijing, 100101, P. R. China
| | - Linqiang Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Chinese Academy of Sciences, Center for Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Kunming, 650223, P. R. China
| | - Yunhai Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Chinese Academy of Sciences, Center for Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Kunming, 650223, P. R. China
| | - Xiaotong Zhu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Institute of Biophysics, Beijing, 100101, P. R. China.,Academy of Sciences, University of Chinese, Beijing, 100049, P. R. China
| | - Shimeng Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Institute of Biophysics, Beijing, 100101, P. R. China.,Academy of Sciences, University of Chinese, Beijing, 100049, P. R. China
| | - Xiao-Ming Zhou
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Institute of Biophysics, Beijing, 100101, P. R. China
| | - Haizhen Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Chinese Academy of Sciences, Center for Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Kunming, 650223, P. R. China
| | - Hongchao Zhang
- General Hospital of Air Force, Beijing, 100142, P. R. China
| | - Bin Liang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan province, Chinese Academy of Sciences, Center for Excellence in Animal Evolution and Genetics, Kunming Institute of Zoology, Kunming, 650223, P. R. China
| | - Pingsheng Liu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Institute of Biophysics, Beijing, 100101, P. R. China.,Academy of Sciences, University of Chinese, Beijing, 100049, P. R. China
| |
Collapse
|
50
|
Jackson CL. Lipid droplet biogenesis. Curr Opin Cell Biol 2019; 59:88-96. [PMID: 31075519 DOI: 10.1016/j.ceb.2019.03.018] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/24/2019] [Accepted: 03/29/2019] [Indexed: 11/18/2022]
Abstract
Lipid droplets (LDs) store neutral lipids in their core as an energy source when nutrients are scarce. The center of an LD is hydrophobic, and hence it is surrounded by a phospholipid monolayer, unlike other organelles that have an aqueous interior and are bounded by a phospholipid bilayer. LDs arise from the ER, where neutral lipid synthesis enzymes are localized. A combination of biophysical analysis and modeling, in vitro reconstitution and cell biological analyses has provided a great deal of information over the past few years on the process of LD biogenesis from the ER. In addition to lipid composition, four protein families (seipin proteins, perilipins, FIT proteins and ER shaping proteins) are crucial for LD biogenesis. Recent studies have shown that LDs preferentially arise, along with peroxisomes, at special ER sites marked by the reticulon-like Pex30/MCTP2 protein. New functions for perilipins and FIT family proteins have been uncovered, and the cryo-electron microscopy structure of seipin coupled with high resolution imaging in cells has provided a more comprehensive picture of its function in LD biogenesis. Seipin, along with other proteins such as Rab18 and its effector NRZ, have been shown to carry out their functions at least in part through regulation of ER-LD contact sites, whose establishment and maintenance have emerged as an essential component of LD biogenesis and maturation.
Collapse
Affiliation(s)
- Catherine L Jackson
- Institut Jacques Monod, UMR7592 CNRS Université Paris-Diderot, Sorbonne Paris Cité, Paris, France.
| |
Collapse
|