1
|
Biswas P, Bako JA, Liston JB, Yu H, Wat LW, Miller CJ, Gordon MD, Huan T, Stanley M, Rideout EJ. Insulin/insulin-like growth factor signaling pathway promotes higher fat storage in Drosophila females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.623936. [PMID: 40342968 PMCID: PMC12060994 DOI: 10.1101/2024.11.18.623936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
In Drosophila , adult females store more fat than males. While the mechanisms that restrict body fat in males are becoming clearer, less is known about how females achieve higher fat storage. Here, we perform a detailed investigation of the mechanisms that promote higher fat storage in females. We show greater intake of dietary sugar supports higher fat storage due to female-biased remodeling of the fat body lipidome. Dietary sugar stimulates a female-specific increase in Drosophila insulin-like peptide 3 (Dilp3), which acts together with greater peripheral insulin sensitivity to augment insulin/insulin-like growth factor signaling pathway (IIS) activity in adult females. Indeed, Dilp3 overexpression prevented the female-biased decrease in body fat after removal of dietary sugar. Given that adult-specific IIS inhibition caused a female-biased decrease in body fat, our data reveal IIS as a key determinant of female fat storage.
Collapse
|
2
|
Toprak U, İnak E, Nauen R. Lipid Metabolism as a Target Site in Pest Control. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39466572 DOI: 10.1007/5584_2024_822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Lipid metabolism is essential to insect life as insects use lipids for their development, reproduction, flight, diapause, and a wide range of other functions. The central organ for insect lipid metabolism is the fat body, which is analogous to mammalian adipose tissue and liver, albeit less structured. Various other systems including the midgut, brain, and neural organs also contribute functionally to insect lipid metabolism. Lipid metabolism is under the control of core lipogenic [e.g. acetyl-CoA-carboxylase (ACC), fatty acid synthase (FAS), perilipin 2 (LSD2)], and lipolytic (lipases, perilipin 1) enzymes that are primarily expressed in the fat body, as well as hormones [insulin-like peptides (ILP), adipokinetic hormone (AKH)], transcription factors (SREBPs, foxO, and CREB), secondary messengers (calcium) and post-translational modifications (phosphorylation). Essential roles of the fat body, together with the fact that proper coordination of lipid metabolism is critical for insects, render lipid metabolism an attractive target site in pest control. In the current chapter, we focus on pest control tactics that target insect lipid metabolism. Various classes of traditional chemical insecticides [e.g. organophosphates, pyrethroids, neonicotinoids, and chitin synthesis inhibitors (Sects. 2.1 and 2.2)] have been shown to interfere with lipid metabolism, albeit it is not their primary site of action. However, the discovery of "lipid biosynthesis inhibitors", tetronic and tetramic acid derivatives commonly known as ketoenols (Sect. 2.3), was a milestone in applied entomology as they directly target lipid biosynthesis, particularly in sucking pests. Spirodiclofen, spiromesifen, and spirotetramat targeting ACC act against various insect and mite pests, while spiropidion and spidoxamat have been introduced to the market only recently. Efforts have concentrated on the development of chemical alternatives, such as hormone agonists and antagonists (Sect. 2.4), dsRNA-based pesticides that depend on RNA interference, which have great potential in pest control (Sect. 2.5) and other eco-friendly alternatives (Sect. 2.6).
Collapse
Affiliation(s)
- Umut Toprak
- Faculty of Agriculture, Department of Plant Protection Ankara, Molecular Entomology Lab, Ankara University, Ankara, Turkey.
| | - Emre İnak
- Faculty of Agriculture, Department of Plant Protection Ankara, Molecular Entomology Lab, Ankara University, Ankara, Turkey
| | - Ralf Nauen
- Bayer AG, Crop Science Division, Monheim, Germany.
| |
Collapse
|
3
|
Henne WM, Ugrankar-Banerjee R, Tran S, Bowerman J, Paul B, Zacharias L, Mathews T, DeBerardinis R. Metabolic rewiring in fat-depleted Drosophila reveals triglyceride:glycogen crosstalk and identifies cDIP as a new regulator of energy metabolism. RESEARCH SQUARE 2024:rs.3.rs-4505077. [PMID: 39483909 PMCID: PMC11527204 DOI: 10.21203/rs.3.rs-4505077/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Tissues store excess nutrients as triglyceride or glycogen, but how these reserves are sensed and communicate remains poorly understood. Here we identify molecular players orchestrating this metabolic balance during fat depletion. We show fat body (FB)-specific depletion of fatty acyl-CoA synthase FASN1 in Drosophila causes near-complete fat loss and metabolic remodeling that dramatically elevates glycogen storage and carbohydrate metabolism. Proteomics and metabolomics identify key factors necessary for rewiring including glycolysis enzymes and target-of-brain-insulin (tobi). FASN1-deficient flies are viable but starvation sensitive, oxidatively stressed, and infertile. We also identify CG10824/cDIP as upregulated in FASN1-depleted Drosophila. cDIP is a leucine-rich-repeat protein with homology to secreted adipokines that fine-tune energy signaling, and is required for fly development in the absence of FASN1. Collectively, we show fat-depleted Drosophila rewire their metabolism to complete development, and identify cDIP as a putative new cytokine that signals fat insufficiency and may regulate energy homeostasis.
Collapse
|
4
|
Wang T, Xu D, Chang X, MacIsaac HJ, Li J, Xu J, Zhang J, Zhang H, Zhou Y, Xu R. Can a shift in dominant species of Microcystis alter growth and reproduction of waterfleas? HARMFUL ALGAE 2024; 136:102657. [PMID: 38876528 DOI: 10.1016/j.hal.2024.102657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024]
Abstract
The bloom-forming species Microcystis wesenbergii and M. aeruginosa occur in many lakes globally, and may exhibit alternating blooms both spatially and temporally. As environmental changes increase, cyanobacteria bloom in more and more lakes and are often dominated by M. wesenbergii. The adverse impact of M. aeruginosa on co-existing organisms including zooplanktonic species has been well-studied, whereas studies of M. wesenbergii are limited. To compare effects of these two species on zooplankton, we explored effects of exudates from different strains of microcystin-producing M. aeruginosa (Ma905 and Ma526) and non-microcystin-producing M. wesenbergii (Mw908 and Mw929), on reproduction by the model zooplankter Daphnia magna in both chronic and acute exposure experiments. Specifically, we tested physiological, biochemical, molecular and transcriptomic characteristics of D. magna exposed to Microcystis exudates. We observed that body length and egg and offspring number of the daphnid increased in all treatments. Among the four strains tested, Ma526 enhanced the size of the first brood, as well as total egg and offspring number. Microcystis exudates stimulated expression of specific genes that induced ecdysone, juvenile hormone, triacylglycerol and vitellogenin biosynthesis, which, in turn, enhanced egg and offspring production of D. magna. Even though all strains of Microcystis affected growth and reproduction, large numbers of downregulated genes involving many essential pathways indicated that the Ma905 strain might contemporaneously induce damage in D. magna. Our study highlights the necessity of including M. wesenbergii into the ecological risk evaluation of cyanobacteria blooms, and emphasizes that consequences to zooplankton may not be clear-cut when assessments are based upon production of microcystins alone.
Collapse
Affiliation(s)
- Tao Wang
- Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, School of Ecology and Environmental Science, Yunnan University, Kunming 650091, China; Ningbo Yonghuanyuan Environmental Engineering and Technology CO., LTD, Ningbo 315000, China
| | - Daochun Xu
- Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, School of Ecology and Environmental Science, Yunnan University, Kunming 650091, China
| | - Xuexiu Chang
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, College of Agronomy and Life Sciences, Kunming University, Kunming 650214, China; Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON N9B 3P4, Canada
| | - Hugh J MacIsaac
- Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, School of Ecology and Environmental Science, Yunnan University, Kunming 650091, China; Great Lakes Institute for Environmental Research, University of Windsor, Windsor, ON N9B 3P4, Canada
| | - Jingjing Li
- Yunnan Collaborative Innovation Center for Plateau Lake Ecology and Environmental Health, College of Agronomy and Life Sciences, Kunming University, Kunming 650214, China
| | - Jun Xu
- Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, School of Ecology and Environmental Science, Yunnan University, Kunming 650091, China
| | - Jinlong Zhang
- Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, School of Ecology and Environmental Science, Yunnan University, Kunming 650091, China
| | - Hongyan Zhang
- Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, School of Ecology and Environmental Science, Yunnan University, Kunming 650091, China
| | - Yuan Zhou
- The Ecological and Environmental Monitoring Station of DEEY in Kunming, Kunming 650228, China
| | - Runbing Xu
- Yunnan Key Laboratory for Plateau Mountain Ecology and Restoration of Degraded Environments, School of Ecology and Environmental Science, Yunnan University, Kunming 650091, China.
| |
Collapse
|
5
|
Chao CF, Pesch YY, Yu H, Wang C, Aristizabal MJ, Huan T, Tanentzapf G, Rideout E. An important role for triglyceride in regulating spermatogenesis. eLife 2024; 12:RP87523. [PMID: 38805376 PMCID: PMC11132686 DOI: 10.7554/elife.87523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Drosophila is a powerful model to study how lipids affect spermatogenesis. Yet, the contribution of neutral lipids, a major lipid group which resides in organelles called lipid droplets (LD), to sperm development is largely unknown. Emerging evidence suggests LD are present in the testis and that loss of neutral lipid- and LD-associated genes causes subfertility; however, key regulators of testis neutral lipids and LD remain unclear. Here, we show LD are present in early-stage somatic and germline cells within the Drosophila testis. We identified a role for triglyceride lipase brummer (bmm) in regulating testis LD, and found that whole-body loss of bmm leads to defects in sperm development. Importantly, these represent cell-autonomous roles for bmm in regulating testis LD and spermatogenesis. Because lipidomic analysis of bmm mutants revealed excess triglyceride accumulation, and spermatogenic defects in bmm mutants were rescued by genetically blocking triglyceride synthesis, our data suggest that bmm-mediated regulation of triglyceride influences sperm development. This identifies triglyceride as an important neutral lipid that contributes to Drosophila sperm development, and reveals a key role for bmm in regulating testis triglyceride levels during spermatogenesis.
Collapse
Affiliation(s)
- Charlotte F Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Yanina-Yasmin Pesch
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Huaxu Yu
- Department of Chemistry, The University of British ColumbiaVancouverCanada
| | - Chenjingyi Wang
- Department of Chemistry, The University of British ColumbiaVancouverCanada
| | | | - Tao Huan
- Department of Chemistry, The University of British ColumbiaVancouverCanada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Elizabeth Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| |
Collapse
|
6
|
Yang F, Xu X, Hu B, Zhang Z, Chen K, Yu Y, Bai H, Tan A. Lipid homeostasis is essential for oogenesis and embryogenesis in the silkworm, Bombyx mori. Cell Mol Life Sci 2024; 81:127. [PMID: 38472536 DOI: 10.1007/s00018-024-05173-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/20/2024] [Accepted: 02/10/2024] [Indexed: 03/14/2024]
Abstract
Reproduction, a fundamental feature of all known life, closely correlates with energy homeostasis. The control of synthesizing and mobilizing lipids are dynamic and well-organized processes to distribute lipid resources across tissues or generations. However, how lipid homeostasis is precisely coordinated during insect reproductive development is poorly understood. Here we describe the relations between energy metabolism and reproduction in the silkworm, Bombyx mori, a lepidopteran model insect, by using CRISPR/Cas9-mediated mutation analysis and comprehensively functional investigation on two major lipid lipases of Brummer (BmBmm) and hormone-sensitive lipase (BmHsl), and the sterol regulatory element binding protein (BmSrebp). BmBmm is a crucial regulator of lipolysis to maintain female fecundity by regulating the triglyceride (TG) storage among the midgut, the fat body, and the ovary. Lipidomics analysis reveals that defective lipolysis of females influences the composition of TG and other membrane lipids in the BmBmm mutant embryos. In contrast, BmHsl mediates embryonic development by controlling sterol metabolism rather than TG metabolism. Transcriptome analysis unveils that BmBmm deficiency significantly improves the expression of lipid synthesis-related genes including BmSrebp in the fat body. Subsequently, we identify BmSrebp as a key regulator of lipid accumulation in oocytes, which promotes oogenesis and cooperates with BmBmm to support the metabolic requirements of oocyte production. In summary, lipid homeostasis plays a vital role in supporting female reproductive success in silkworms.
Collapse
Affiliation(s)
- Fangying Yang
- Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Xiaoyan Xu
- Core Facility Center, CAS Center for Excellence in Molecular Plant Sciences, Chinese Academy of Sciences, Shanghai, 200032, China
| | - Bo Hu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Zhongjie Zhang
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Kai Chen
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Ye Yu
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China
| | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, IA, 50011, USA
| | - Anjiang Tan
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, 212100, China.
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, The Sericultural Research Institute, Chinese Academy of Agricultural Sciences, Zhenjiang, 212100, China.
| |
Collapse
|
7
|
Berg C, Sieber M, Sun J. Finishing the egg. Genetics 2024; 226:iyad183. [PMID: 38000906 PMCID: PMC10763546 DOI: 10.1093/genetics/iyad183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/27/2023] [Indexed: 11/26/2023] Open
Abstract
Gamete development is a fundamental process that is highly conserved from early eukaryotes to mammals. As germ cells develop, they must coordinate a dynamic series of cellular processes that support growth, cell specification, patterning, the loading of maternal factors (RNAs, proteins, and nutrients), differentiation of structures to enable fertilization and ensure embryonic survival, and other processes that make a functional oocyte. To achieve these goals, germ cells integrate a complex milieu of environmental and developmental signals to produce fertilizable eggs. Over the past 50 years, Drosophila oogenesis has risen to the forefront as a system to interrogate the sophisticated mechanisms that drive oocyte development. Studies in Drosophila have defined mechanisms in germ cells that control meiosis, protect genome integrity, facilitate mRNA trafficking, and support the maternal loading of nutrients. Work in this system has provided key insights into the mechanisms that establish egg chamber polarity and patterning as well as the mechanisms that drive ovulation and egg activation. Using the power of Drosophila genetics, the field has begun to define the molecular mechanisms that coordinate environmental stresses and nutrient availability with oocyte development. Importantly, the majority of these reproductive mechanisms are highly conserved throughout evolution, and many play critical roles in the development of somatic tissues as well. In this chapter, we summarize the recent progress in several key areas that impact egg chamber development and ovulation. First, we discuss the mechanisms that drive nutrient storage and trafficking during oocyte maturation and vitellogenesis. Second, we examine the processes that regulate follicle cell patterning and how that patterning impacts the construction of the egg shell and the establishment of embryonic polarity. Finally, we examine regulatory factors that control ovulation, egg activation, and successful fertilization.
Collapse
Affiliation(s)
- Celeste Berg
- Department of Genome Sciences, University of Washington, Seattle, WA 98195-5065 USA
| | - Matthew Sieber
- Department of Physiology, UT Southwestern Medical Center, Dallas, TX 75390 USA
| | - Jianjun Sun
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT 06269 USA
| |
Collapse
|
8
|
Kim N, Ahn Y, Ko K, Kim B, Han K, Suh HJ, Jung J, Hong KB. Yeast Hydrolysate Inhibits Lipid Accumulation via Regulation of Lipid Accumulation-Related Genes in a Drosophila Model of High-Sugar Diet-Induced Obesity. Int J Mol Sci 2023; 24:16302. [PMID: 38003491 PMCID: PMC10671767 DOI: 10.3390/ijms242216302] [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: 09/25/2023] [Revised: 10/31/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
The increasing frequency of processed food consumption has led to the higher ingestion of sugar, increasing the risk of chronic diseases, such as obesity. Yeast hydrolysates (YHs) inhibit body fat accumulation. However, the action mechanism of YH in relation to high-sugar diet-induced obesity is still unclear. Therefore, this study aimed to evaluate the biological effects of YH on lipid accumulation and verify behavioral changes and carbohydrate metabolic gene regulation in high-sugar diet-fed fruit flies. Adult male flies (Drosophila melanogaster; 2-5 days old) were exposed to 20% sucrose for obesity induction. In high-sugar-fed Drosophila, the effect of YH was compared with that of yeast extract. The effects of YH on body conditions and lipid droplet size were quantified and analyzed. Behavioral factors were evaluated by analyzing circadian rhythm patterns and neurotransmitter content, and a molecular approach was used to analyze the expression of metabolism-related genes. Dietary supplementation with YH did not reduce total sugar content, but significantly decreased the triglyceride (TG) levels in Drosophila. A behavioral analysis showed that the total number of night-time activities increased significantly with YH treatment in a dose-dependent manner. In addition, YH effectively regulated the gene expression of insulin-like peptides related to carbohydrate metabolism as well as genes related to lipogenesis. The TG content was significantly reduced at a YH concentration of 0.5%, confirming that the active compound in YH effectively suppresses fat accumulation. These findings support that YH is a potential anti-obesity food material via regulating carbohydrate metabolism in Drosophila.
Collapse
Affiliation(s)
- Nari Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (K.H.); (H.J.S.)
| | - Yejin Ahn
- Research Group of Functional Food Materials, Korea Food Research Institute, Wanju 55365, Republic of Korea;
| | - Kayoung Ko
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea;
| | - Boyun Kim
- Department of SmartBio, Kyungsung University, Busan 48434, Republic of Korea;
| | - Kisoo Han
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (K.H.); (H.J.S.)
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul 02841, Republic of Korea; (N.K.); (K.H.); (H.J.S.)
| | - Jewon Jung
- Department of SmartBio, Kyungsung University, Busan 48434, Republic of Korea;
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju 63243, Republic of Korea;
| |
Collapse
|
9
|
Giedt MS, Thomalla JM, White RP, Johnson MR, Lai ZW, Tootle TL, Welte MA. Adipose triglyceride lipase promotes prostaglandin-dependent actin remodeling by regulating substrate release from lipid droplets. Development 2023; 150:dev201516. [PMID: 37306387 PMCID: PMC10281261 DOI: 10.1242/dev.201516] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 05/03/2023] [Indexed: 05/13/2023]
Abstract
Lipid droplets (LDs), crucial regulators of lipid metabolism, accumulate during oocyte development. However, their roles in fertility remain largely unknown. During Drosophila oogenesis, LD accumulation coincides with the actin remodeling necessary for follicle development. Loss of the LD-associated Adipose Triglyceride Lipase (ATGL) disrupts both actin bundle formation and cortical actin integrity, an unusual phenotype also seen when the prostaglandin (PG) synthase Pxt is missing. Dominant genetic interactions and PG treatment of follicles indicate that ATGL acts upstream of Pxt to regulate actin remodeling. Our data suggest that ATGL releases arachidonic acid (AA) from LDs to serve as the substrate for PG synthesis. Lipidomic analysis detects AA-containing triglycerides in ovaries, and these are increased when ATGL is lost. High levels of exogenous AA block follicle development; this is enhanced by impairing LD formation and suppressed by reducing ATGL. Together, these data support the model that AA stored in LD triglycerides is released by ATGL to drive the production of PGs, which promote the actin remodeling necessary for follicle development. We speculate that this pathway is conserved across organisms to regulate oocyte development and promote fertility.
Collapse
Affiliation(s)
- Michelle S. Giedt
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | | | - Roger P. White
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Matthew R. Johnson
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| | - Zon Weng Lai
- Harvard T.H. Chan Advanced Multi-omics Platform, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tina L. Tootle
- Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Michael A. Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, USA
| |
Collapse
|
10
|
Tippetts TS, Sieber MH, Solmonson A. Beyond energy and growth: the role of metabolism in developmental signaling, cell behavior and diapause. Development 2023; 150:dev201610. [PMID: 37883062 PMCID: PMC10652041 DOI: 10.1242/dev.201610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
Metabolism is crucial for development through supporting cell growth, energy production, establishing cell identity, developmental signaling and pattern formation. In many model systems, development occurs alongside metabolic transitions as cells differentiate and specialize in metabolism that supports new functions. Some cells exhibit metabolic flexibility to circumvent mutations or aberrant signaling, whereas other cell types require specific nutrients for developmental progress. Metabolic gradients and protein modifications enable pattern formation and cell communication. On an organism level, inadequate nutrients or stress can limit germ cell maturation, implantation and maturity through diapause, which slows metabolic activities until embryonic activation under improved environmental conditions.
Collapse
Affiliation(s)
- Trevor S. Tippetts
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Matthew H. Sieber
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ashley Solmonson
- Laboratory of Developmental Metabolism and Placental Biology, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
11
|
Roy SD, Nagarajan S, Jalal MS, Basar MA, Duttaroy A. New mutant alleles for Spargel/dPGC-1 highlights the function of Spargel RRM domain in oogenesis and expands the role of Spargel in embryogenesis and intracellular transport. G3 (BETHESDA, MD.) 2023; 13:jkad142. [PMID: 37369430 PMCID: PMC10468312 DOI: 10.1093/g3journal/jkad142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 01/24/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023]
Abstract
Energy metabolism in vertebrates is controlled by three members of the PGC-1 (PPAR γ- coactivator 1) family, transcriptional coactivators that shape responses to physiological stimuli by interacting with the nuclear receptors and other transcription factors. Multiple evidence now supports that Spargel protein found in insects and ascidians is the ancestral form of vertebrate PGC-1's. Here, we undertook functional analysis of srl gene in Drosophila, asking about the requirement of Spargel per se during embryogenesis and its RNA binding domains. CRISPR- engineered srl gene deletion turned out to be an amorphic allele that is late embryonic/early larval lethal and Spargel protein missing its RNA binding domain (SrlΔRRM) negatively affects female fertility. Overexpression of wild-type Spargel in transgenic flies expedited the growth of egg chambers. On the other hand, oogenesis is blocked in a dominant-negative fashion in the presence of excess Spargel lacking its RRM domains. Finally, we observed aggregation of Notch proteins in egg chambers of srl mutant flies, suggesting that Spargel is involved in intracellular transport of Notch proteins. Taken together, we claim that these new mutant alleles of spargel are emerging powerful tools for revealing new biological functions for Spargel, an essential transcription coactivator in both Drosophila and mammals.
Collapse
Affiliation(s)
- Swagota D Roy
- Biology Department, Howard University, 415 College St. NW, Washington D.C., USA 20059
| | - Sabarish Nagarajan
- Biology Department, Howard University, 415 College St. NW, Washington D.C., USA 20059
| | - Md Shah Jalal
- Biology Department, Howard University, 415 College St. NW, Washington D.C., USA 20059
| | - Md Abul Basar
- Biology Department, Howard University, 415 College St. NW, Washington D.C., USA 20059
| | - Atanu Duttaroy
- Biology Department, Howard University, 415 College St. NW, Washington D.C., USA 20059
| |
Collapse
|
12
|
Sun R, Jiang L, Chen W, Xu Y, Yi X, Zhong G. Azadirachtin exposure inhibit ovary development of Spodoptera litura (Lepidoptera: Noctuidae) by altering lipids metabolism event and inhibiting insulin signaling pathways. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 262:115151. [PMID: 37356396 DOI: 10.1016/j.ecoenv.2023.115151] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/27/2023]
Abstract
Lipids are main energy source for insects reproduction, which are becoming emerging target for pest management. Azadirachtin (AZA) is a multi-targeted and promising botanical insecticide, but its reproduction toxicity mechanism related to lipids metabolism is poorly understood. Here, we applied lipidomic and transcriptomic to provide a comprehensive resource for describing the effect of AZA on lipids remodeling in ovary of Spodoptera litura. The results showed that AZA exposure obviously altered the contents of 130 lipids subclasses (76 upregulated and 54 downregulated). In detail, AZA exposure changed the length and saturation degrees of fatty acyl chain of most glycerolipid, phospholipid and sphingolipid as well as the expression of genes related to biosynthesis of unsaturated fatty acids and fatty acids elongation. Besides, following the abnormal lipids metabolism, western blot analysis suggested that AZA induce insulin resistance-like phenotypes by inhibiting insulin receptor substrates (IRS) /PI3K/AKT pathway, which might be responsible for the ovary abnormalities of S. litura. Collectively, our study provided insights into the lipids metabolism event in S. litura underlying AZA exposure, these key metabolites and genes identified in this study would also provide important reference for pest control in future.
Collapse
Affiliation(s)
- Ranran Sun
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou, China; Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Liwei Jiang
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou, China; Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Wenlong Chen
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou, China; Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Yuanhao Xu
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou, China; Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China
| | - Xin Yi
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou, China; Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China.
| | - Guohua Zhong
- Key Laboratory of Crop Integrated Pest Management in South China, Ministry of Agriculture, South China Agricultural University, Guangzhou, China; Key Laboratory of Natural Pesticide and Chemical Biology, Ministry of Education, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
13
|
Wolf S, Abhyankar V, Melo D, Ayroles JF, Pallares LF. From GWAS to signal validation: An approach for estimating genetic effects while preserving genomic context. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.09.531909. [PMID: 36945453 PMCID: PMC10028994 DOI: 10.1101/2023.03.09.531909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Validating associations between genotypic and phenotypic variation remains a challenge, despite advancements in association studies. Common approaches for signal validation rely on gene-level perturbations, such as loss-of-function mutations or RNAi, which test the effect of genetic modifications usually not observed in nature. CRISPR-based methods can validate associations at the SNP level, but have significant drawbacks, including resulting off-target effects and being both time-consuming and expensive. Both approaches usually modify the genome of a single genetic background, limiting the generalizability of experiments. To address these challenges, we present a simple, low-cost experimental scheme for validating genetic associations at the SNP level in outbred populations. The approach involves genotyping live outbred individuals at a focal SNP, crossing homozygous individuals with the same genotype at that locus, and contrasting phenotypes across resulting synthetic outbred populations. We tested this method in Drosophila melanogaster, measuring the longevity effects of a polymorphism at a naturally-segregating cis-eQTL for the midway gene. Our results demonstrate the utility of this method in SNP-level validation of naturally occurring genetic variation regulating complex traits. This method provides a bridge between the statistical discovery of genotype-phenotype associations and their validation in the natural context of heterogeneous genomic contexts.
Collapse
Affiliation(s)
- Scott Wolf
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Varada Abhyankar
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Diogo Melo
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Julien F. Ayroles
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
| | - Luisa F. Pallares
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ, USA
- Current address: Friedrich Miescher Laboratory of the Max Planck Society, Tübingen, Germany
| |
Collapse
|
14
|
Bresgen N, Kovacs M, Lahnsteiner A, Felder TK, Rinnerthaler M. The Janus-Faced Role of Lipid Droplets in Aging: Insights from the Cellular Perspective. Biomolecules 2023; 13:912. [PMID: 37371492 PMCID: PMC10301655 DOI: 10.3390/biom13060912] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
It is widely accepted that nine hallmarks-including mitochondrial dysfunction, epigenetic alterations, and loss of proteostasis-exist that describe the cellular aging process. Adding to this, a well-described cell organelle in the metabolic context, namely, lipid droplets, also accumulates with increasing age, which can be regarded as a further aging-associated process. Independently of their essential role as fat stores, lipid droplets are also able to control cell integrity by mitigating lipotoxic and proteotoxic insults. As we will show in this review, numerous longevity interventions (such as mTOR inhibition) also lead to strong accumulation of lipid droplets in Saccharomyces cerevisiae, Caenorhabditis elegans, Drosophila melanogaster, and mammalian cells, just to name a few examples. In mammals, due to the variety of different cell types and tissues, the role of lipid droplets during the aging process is much more complex. Using selected diseases associated with aging, such as Alzheimer's disease, Parkinson's disease, type II diabetes, and cardiovascular disease, we show that lipid droplets are "Janus"-faced. In an early phase of the disease, lipid droplets mitigate the toxicity of lipid peroxidation and protein aggregates, but in a later phase of the disease, a strong accumulation of lipid droplets can cause problems for cells and tissues.
Collapse
Affiliation(s)
- Nikolaus Bresgen
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Melanie Kovacs
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Angelika Lahnsteiner
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| | - Thomas Klaus Felder
- Department of Laboratory Medicine, Paracelsus Medical University, 5020 Salzburg, Austria
| | - Mark Rinnerthaler
- Department of Biosciences and Medical Biology, Paris-Lodron University Salzburg, 5020 Salzburg, Austria; (N.B.)
| |
Collapse
|
15
|
Zhao T, Wang M, Li Z, Li H, Yuan D, Zhang X, Guo M, Qian W, Cheng D. Wds-Mediated H3K4me3 Modification Regulates Lipid Synthesis and Transport in Drosophila. Int J Mol Sci 2023; 24:ijms24076125. [PMID: 37047100 PMCID: PMC10093852 DOI: 10.3390/ijms24076125] [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: 02/14/2023] [Revised: 03/10/2023] [Accepted: 03/16/2023] [Indexed: 04/14/2023] Open
Abstract
Lipid homeostasis is essential for insect growth and development. The complex of proteins associated with Set 1 (COMPASS)-catalyzed Histone 3 lysine 4 trimethylation (H3K4me3) epigenetically activates gene transcription and is involved in various biological processes, but the role and molecular mechanism of H3K4me3 modification in lipid homeostasis remains largely unknown. In the present study, we showed in Drosophila that fat body-specific knockdown of will die slowly (Wds) as one of the COMPASS complex components caused a decrease in lipid droplet (LD) size and triglyceride (TG) levels. Mechanistically, Wds-mediated H3K4me3 modification in the fat body targeted several lipogenic genes involved in lipid synthesis and the Lpp gene associated with lipid transport to promote their expressions; the transcription factor heat shock factor (Hsf) could interact with Wds to modulate H3K4me3 modification within the promoters of these targets; and fat body-specific knockdown of Hsf phenocopied the effects of Wds knockdown on lipid homeostasis in the fat body. Moreover, fat body-specific knockdown of Wds or Hsf reduced high-fat diet (HFD)-induced oversized LDs and high TG levels. Altogether, our study reveals that Wds-mediated H3K4me3 modification is required for lipid homeostasis during Drosophila development and provides novel insights into the epigenetic regulation of insect lipid metabolism.
Collapse
Affiliation(s)
- Tujing Zhao
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Min Wang
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Zheng Li
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Hao Li
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Dongqin Yuan
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Xing Zhang
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Mengge Guo
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Wenliang Qian
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| | - Daojun Cheng
- State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Sericultural Science, Southwest University, Chongqing 400715, China
| |
Collapse
|
16
|
Abstract
In this chapter, we highlight examples of the diverse array of developmental, cellular, and biochemical insights that can be gained by using Drosophila melanogaster oogenesis as a model tissue. We begin with an overview of ovary development and adult oogenesis. Then we summarize how the adult Drosophila ovary continues to advance our understanding of stem cells, cell cycle, cell migration, cytoplasmic streaming, nurse cell dumping, and cell death. We also review emerging areas of study, including the roles of lipid droplets, ribosomes, and nuclear actin in egg development. Finally, we conclude by discussing the growing conservation of processes and signaling pathways that regulate oogenesis and female reproduction from flies to humans.
Collapse
|
17
|
White RP, Welte MA. Visualizing Lipid Droplets in Drosophila Oogenesis. Methods Mol Biol 2023; 2626:233-251. [PMID: 36715908 DOI: 10.1007/978-1-0716-2970-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Lipid droplets (LDs) are fat storage organelles highly abundant in oocytes and eggs of many vertebrates and invertebrates. They have roles both during oogenesis and in provisioning the developing embryo. In Drosophila, large numbers of LDs are generated in nurse cells during mid-oogenesis and then transferred to oocytes. Their number and spatial distribution changes developmentally and in response to various experimental manipulations. This chapter demonstrates how to visualize LDs in Drosophila follicles, both in fixed tissues and living samples. For fixed samples, the protocol explains how to prepare female flies, dissect ovaries, isolate follicles, fix, apply stains, mount the tissue, and perform imaging. For live samples, the protocol shows how to dissect ovaries, apply a fluorescent LD dye, and culture follicles such that they remain alive and healthy during imaging. Finally, a method is provided that employs in vivo centrifugation to assess colocalization of markers with LDs.
Collapse
Affiliation(s)
- Roger P White
- Department of Biology, University of Rochester, Rochester, NY, USA
| | - Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
18
|
Pallares LF, Lea AJ, Han C, Filippova EV, Andolfatto P, Ayroles JF. Dietary stress remodels the genetic architecture of lifespan variation in outbred Drosophila. Nat Genet 2023; 55:123-129. [PMID: 36550361 DOI: 10.1038/s41588-022-01246-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/26/2022] [Indexed: 12/24/2022]
Abstract
Evolutionary theory suggests that lifespan-reducing alleles should be purged from the gene pool, and yet decades of genome-wide association and model organism studies have shown that they persist. One potential explanation is that alleles that regulate lifespan do so only in certain environmental contexts. We exposed outbred Drosophila to control and high-sugar diets and genotyped more than 10,000 adult flies to track allele frequency changes over the course of a single adult lifespan. We identified thousands of lifespan-associated alleles associated with early versus late-life trade-offs, late-onset effects and genotype-by-environment interactions. Remarkably, a third of lifespan-associated genetic variation had environmentally dependent effects on lifespan. We find that lifespan-reducing alleles are often recently derived, have stronger effects on a high-sugar diet and show signatures of selection in wild Drosophila populations, consistent with the evolutionary mismatch hypothesis. Our results provide insight into the highly polygenic and context-dependent genetic architecture of lifespan variation and the evolutionary processes that shape this key trait.
Collapse
Affiliation(s)
- Luisa F Pallares
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ecology and Evolutionary Biology Department, Princeton University, Princeton, NJ, USA
- Friedrich Miescher Laboratory, Max Planck Society, Tübingen, Germany
| | - Amanda J Lea
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ecology and Evolutionary Biology Department, Princeton University, Princeton, NJ, USA
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Clair Han
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Janelia Research Campus of the Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Elena V Filippova
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Peter Andolfatto
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Julien F Ayroles
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA.
- Ecology and Evolutionary Biology Department, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
19
|
Zhang S, Xie J, Luo R, Zhang H, Zheng W. MiR-2b-2-5p regulates lipid metabolism and reproduction by targeting CREB in Bactrocera dorsalis. RNA Biol 2023; 20:164-176. [PMID: 37092804 PMCID: PMC10128458 DOI: 10.1080/15476286.2023.2204579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2023] Open
Abstract
In female animals, metabolic homoeostasis and reproductive fitness are critical to population expansion. The trade-off between lipid storage and reproduction inevitably occurs. However, most studies have focused on the complex network of relationships between reproductive and metabolic physiology at the transcriptional level. In this study, we identified a microRNA, miR-2b-2-5p, in a highly invasive quarantine pest, Bactrocera dorsalis. Knockdown of miR-2b-2-5p by antagomiR microinjection impaired ovarian development, reduced fecundity, and decreased triglyceride (TAG) storage in the fat body, whereas overexpression of miR-2b-2-5p by injection of its mimic caused reproductive defects similar to knockdown but increased TAG. Bioinformatics analysis and dual luciferase assay indicated that cyclic AMP response element (CRE)-binding protein (CREB) was the target gene of miR-2b-2-5p. RNAi-mediated knockdown of CREB led to excessive lipid storage and reproductive defects. Further starvation treatment revealed that miR-2b-2-5p functions by fine-tuning CREB expression in response to dietary stimuli. These results suggest that miR-2b-2-5p acts as a monitor to regulate CREB mRNA levels in the fat body, maintaining lipid homoeostasis and keeping the reproductive system on track. Thus, our study not only provides new insights into the interaction between metabolism and reproduction at the posttranscriptional level in B. dorsalis, but also providing a potential eco-friendly control strategy (RNAi-based biopesticides targeting essential miRNAs) for this notorious agricultural pest.
Collapse
Affiliation(s)
- Shengfeng Zhang
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Junfei Xie
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Rengang Luo
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Hongyu Zhang
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Weiwei Zheng
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, Hubei Hongshan Laboratory, Hubei Key Laboratory of Insect Resource Application and Sustainable Pest Control, Institute of Urban and Horticultural Entomology, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
20
|
Zhou S, Lu Y, Chen J, Pan Z, Pang L, Wang Y, Zhang Q, Strand MR, Chen XX, Huang J. Parasite reliance on its host gut microbiota for nutrition and survival. THE ISME JOURNAL 2022; 16:2574-2586. [PMID: 35941172 PMCID: PMC9561699 DOI: 10.1038/s41396-022-01301-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 11/12/2022]
Abstract
Studying the microbial symbionts of eukaryotic hosts has revealed a range of interactions that benefit host biology. Most eukaryotes are also infected by parasites that adversely affect host biology for their own benefit. However, it is largely unclear whether the ability of parasites to develop in hosts also depends on host-associated symbionts, e.g., the gut microbiota. Here, we studied the parasitic wasp Leptopilina boulardi (Lb) and its host Drosophila melanogaster. Results showed that Lb successfully develops in conventional hosts (CN) with a gut microbiota but fails to develop in axenic hosts (AX) without a gut microbiota. We determined that developing Lb larvae consume fat body cells that store lipids. We also determined that much larger amounts of lipid accumulate in fat body cells of parasitized CN hosts than parasitized AX hosts. CN hosts parasitized by Lb exhibited large increases in the abundance of the bacterium Acetobacter pomorum in the gut, but did not affect the abundance of Lactobacillus fructivorans which is another common member of the host gut microbiota. However, AX hosts inoculated with A. pomorum and/or L. fructivorans did not rescue development of Lb. In contrast, AX larvae inoculated with A. pomorum plus other identified gut community members including a Bacillus sp. substantially rescued Lb development. Rescue was further associated with increased lipid accumulation in host fat body cells. Insulin-like peptides increased in brain neurosecretory cells of parasitized CN larvae. Lipid accumulation in the fat body of CN hosts was further associated with reduced Bmm lipase activity mediated by insulin/insulin-like growth factor signaling (IIS). Altogether, our results identify a previously unknown role for the gut microbiota in defining host permissiveness for a parasite. Our findings also identify a new paradigm for parasite manipulation of host metabolism that depends on insulin signaling and the gut microbiota.
Collapse
Affiliation(s)
- Sicong Zhou
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Yueqi Lu
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Jiani Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Zhongqiu Pan
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Lan Pang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Ying Wang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Qichao Zhang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China
| | - Michael R Strand
- Department of Entomology, University of Georgia, Athens, GA, 30602, USA.
| | - Xue-Xin Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China.
- State Key Lab of Rice Biology, Zhejiang University, Hangzhou, 310058, China.
| | - Jianhua Huang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, 310058, China.
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, 310058, China.
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
21
|
Chen G, Harwood JL, Lemieux MJ, Stone SJ, Weselake RJ. Acyl-CoA:diacylglycerol acyltransferase: Properties, physiological roles, metabolic engineering and intentional control. Prog Lipid Res 2022; 88:101181. [PMID: 35820474 DOI: 10.1016/j.plipres.2022.101181] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 12/15/2022]
Abstract
Acyl-CoA:diacylglycerol acyltransferase (DGAT, EC 2.3.1.20) catalyzes the last reaction in the acyl-CoA-dependent biosynthesis of triacylglycerol (TAG). DGAT activity resides mainly in membrane-bound DGAT1 and DGAT2 in eukaryotes and bifunctional wax ester synthase-diacylglycerol acyltransferase (WSD) in bacteria, which are all membrane-bound proteins but exhibit no sequence homology to each other. Recent studies also identified other DGAT enzymes such as the soluble DGAT3 and diacylglycerol acetyltransferase (EaDAcT), as well as enzymes with DGAT activities including defective in cuticular ridges (DCR) and steryl and phytyl ester synthases (PESs). This review comprehensively discusses research advances on DGATs in prokaryotes and eukaryotes with a focus on their biochemical properties, physiological roles, and biotechnological and therapeutic applications. The review begins with a discussion of DGAT assay methods, followed by a systematic discussion of TAG biosynthesis and the properties and physiological role of DGATs. Thereafter, the review discusses the three-dimensional structure and insights into mechanism of action of human DGAT1, and the modeled DGAT1 from Brassica napus. The review then examines metabolic engineering strategies involving manipulation of DGAT, followed by a discussion of its therapeutic applications. DGAT in relation to improvement of livestock traits is also discussed along with DGATs in various other eukaryotic organisms.
Collapse
Affiliation(s)
- Guanqun Chen
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada.
| | - John L Harwood
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Membrane Protein Disease Research Group, Edmonton T6G 2H7, Canada
| | - Scot J Stone
- Department of Biochemistry, Microbiology and Immunology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 5E5, Canada.
| | - Randall J Weselake
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Alberta T6H 2P5, Canada
| |
Collapse
|
22
|
Nascimento da Silva J, Calixto Conceição C, Cristina Ramos de Brito G, Costa Santos D, Martins da Silva R, Arcanjo A, Henrique Ferreira Sorgine M, de Oliveira PL, Andrade Moreira L, da Silva Vaz I, Logullo C. Wolbachia pipientis modulates metabolism and immunity during Aedes fluviatilis oogenesis. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2022; 146:103776. [PMID: 35526745 DOI: 10.1016/j.ibmb.2022.103776] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/25/2022] [Accepted: 04/25/2022] [Indexed: 06/14/2023]
Abstract
Wolbachia pipientis is a maternally transmitted bacterium that mostly colonizes arthropods, including the mosquito Aedes fluviatilis, potentially affecting different aspects of host physiology. This intracellular bacterium prefers gonadal tissue cells, interfering with the reproductive cycle of insects, arachnids, crustaceans, and nematodes. Wolbachia's ability to modulate the host's reproduction is related to its success in prevalence and frequency. Infecting oocytes is essential for vertical propagation, ensuring its presence in the germline. The mosquito Ae. fluviatilis is a natural host for this bacterium and therefore represents an excellent experimental model in the effort to understand host-symbiont interactions and the mutual metabolic regulation. The aim of this study was to comparatively describe metabolic changes in naturally Wolbachia-infected and uninfected ovaries of Ae. fluviatilis during the vitellogenic period of oogenesis, thus increasing the knowledge about Wolbachia parasitic/symbiotic mechanisms.
Collapse
Affiliation(s)
- Jhenifer Nascimento da Silva
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Christiano Calixto Conceição
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Gisely Cristina Ramos de Brito
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Daniel Costa Santos
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Renato Martins da Silva
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Angélica Arcanjo
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Marcos Henrique Ferreira Sorgine
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Pedro L de Oliveira
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil
| | - Luciano Andrade Moreira
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil; Grupo Mosquitos Vetores: Endossimbiontes e Interação Patógeno Vetor, Instituto René Rachou - Fiocruz, Belo Horizonte, Minas Gerais, Brazil
| | - Itabajara da Silva Vaz
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil; Centro de Biotecnologia and Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Carlos Logullo
- Laboratório de Bioquímica de Artrópodes Hematófagos, Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, RJ, Brazil; Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
23
|
Gáliková M, Klepsatel P. Endocrine control of glycogen and triacylglycerol breakdown in the fly model. Semin Cell Dev Biol 2022; 138:104-116. [PMID: 35393234 DOI: 10.1016/j.semcdb.2022.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 03/15/2022] [Accepted: 03/28/2022] [Indexed: 12/12/2022]
Abstract
Over the last decade, the combination of genetics, transcriptomic and proteomic approaches yielded substantial insights into the mechanisms behind the synthesis and breakdown of energy stores in the model organisms. The fruit fly Drosophila melanogaster has been particularly useful to unravel genetic regulations of energy metabolism. Despite the considerable evolutionary distance between humans and flies, the energy storage organs, main metabolic pathways, and even their genetic regulations remained relatively conserved. Glycogen and fat are universal energy reserves used in all animal phyla and several of their endocrine regulators, such as the insulin pathway, are highly evolutionarily conserved. Nevertheless, some of the factors inducing catabolism of energy stores have diverged significantly during evolution. Moreover, even within a single insect species, D. melanogaster, there are substantial developmental and context-dependent variances in the regulation of energy stores. These differences include, among others, the endocrine pathways that govern the catabolic events or the predominant fuel which is utilized for the given process. For example, many catabolic regulators that control energy reserves in adulthood seem to be largely dispensable for energy mobilization during development. In this review, we focus on a selection of the most important catabolic regulators from the group of peptide hormones (Adipokinetic hormone, Corazonin), catecholamines (octopamine), steroid hormones (20-hydroxyecdysone), and other factors (extracellular adenosine, regulators of lipase Brummer). We discuss their roles in the mobilization of energy reserves for processes such as development through non-feeding stages, flight or starvation survival. Finally, we conclude with future perspectives on the energy balance research in the fly model.
Collapse
Affiliation(s)
- Martina Gáliková
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 06 Bratislava, Slovakia.
| | - Peter Klepsatel
- Institute of Zoology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 06 Bratislava, Slovakia; Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, Dúbravská cesta 9, 840 05 Bratislava, Slovakia
| |
Collapse
|
24
|
Séité S, Harrison MC, Sillam-Dussès D, Lupoli R, Van Dooren TJM, Robert A, Poissonnier LA, Lemainque A, Renault D, Acket S, Andrieu M, Viscarra J, Sul HS, de Beer ZW, Bornberg-Bauer E, Vasseur-Cognet M. Lifespan prolonging mechanisms and insulin upregulation without fat accumulation in long-lived reproductives of a higher termite. Commun Biol 2022; 5:44. [PMID: 35027667 PMCID: PMC8758687 DOI: 10.1038/s42003-021-02974-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/07/2021] [Indexed: 11/30/2022] Open
Abstract
Kings and queens of eusocial termites can live for decades, while queens sustain a nearly maximal fertility. To investigate the molecular mechanisms underlying their long lifespan, we carried out transcriptomics, lipidomics and metabolomics in Macrotermes natalensis on sterile short-lived workers, long-lived kings and five stages spanning twenty years of adult queen maturation. Reproductives share gene expression differences from workers in agreement with a reduction of several aging-related processes, involving upregulation of DNA damage repair and mitochondrial functions. Anti-oxidant gene expression is downregulated, while peroxidability of membranes in queens decreases. Against expectations, we observed an upregulated gene expression in fat bodies of reproductives of several components of the IIS pathway, including an insulin-like peptide, Ilp9. This pattern does not lead to deleterious fat storage in physogastric queens, while simple sugars dominate in their hemolymph and large amounts of resources are allocated towards oogenesis. Our findings support the notion that all processes causing aging need to be addressed simultaneously in order to prevent it.
Collapse
Affiliation(s)
- Sarah Séité
- UMR IRD 242, UPEC, CNRS 7618, UPMC 113, INRAe 1392, Paris 7 113, Institute of Ecology and Environmental Sciences of Paris, Bondy, France
- University of Paris-Est, Créteil, France
| | - Mark C Harrison
- Institute for Evolution and Biodiversity, University of Münster, Münster, Germany
| | - David Sillam-Dussès
- University Sorbonne Paris Nord, Laboratory of Experimental and Comparative Ethology, UR4443, Villetaneuse, France
| | - Roland Lupoli
- UMR IRD 242, UPEC, CNRS 7618, UPMC 113, INRAe 1392, Paris 7 113, Institute of Ecology and Environmental Sciences of Paris, Bondy, France
- University of Paris-Est, Créteil, France
| | - Tom J M Van Dooren
- UMR UPMC 113, IRD 242, UPEC, CNRS 7618, INRA 1392, PARIS 7 113, Institute of Ecology and Environmental Sciences of Paris, Paris, France
- Naturalis Biodiversity Center, Leiden, The Netherlands
| | - Alain Robert
- University Sorbonne Paris Nord, Laboratory of Experimental and Comparative Ethology, UR4443, Villetaneuse, France
| | - Laure-Anne Poissonnier
- Department of Biochemistry, Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Arnaud Lemainque
- Genoscope, François-Jacob Institute of Biology, Alternative Energies and Atomic Energy Commission, University of Paris-Saclay, Evry, France
| | - David Renault
- University of Rennes, CNRS, ECOBIO (Ecosystems, biodiversity, evolution) - UMR, 6553, Rennes, France
- University Institute of France, Paris, France
| | - Sébastien Acket
- University of Technology of Compiègne, UPJV, UMR CNRS 7025, Enzyme and Cell Engineering, Royallieu research Center, Compiègne, France
| | - Muriel Andrieu
- Cochin Institute, UMR INSERM U1016, CNRS 8104, University of Paris Descartes, CYBIO Platform, Paris, France
| | - José Viscarra
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Hei Sook Sul
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA
| | - Z Wilhelm de Beer
- Department of Biochemistry, Genetics and Microbiology, Forestry and Agricultural Biotechnology Institute, University of Pretoria, Pretoria, South Africa
| | - Erich Bornberg-Bauer
- Institute for Evolution and Biodiversity, University of Münster, Münster, Germany
| | - Mireille Vasseur-Cognet
- UMR IRD 242, UPEC, CNRS 7618, UPMC 113, INRAe 1392, Paris 7 113, Institute of Ecology and Environmental Sciences of Paris, Bondy, France.
- University of Paris-Est, Créteil, France.
- INSERM, Paris, France.
| |
Collapse
|
25
|
Russell SA, Laws KM, Bashaw GJ. Frazzled/Dcc acts independently of Netrin to promote germline survival during Drosophila oogenesis. Development 2021; 148:dev199762. [PMID: 34910816 PMCID: PMC8722396 DOI: 10.1242/dev.199762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 11/16/2021] [Indexed: 11/20/2022]
Abstract
The Netrin receptor Frazzled/Dcc (Fra in Drosophila) functions in diverse tissue contexts to regulate cell migration, axon guidance and cell survival. Fra signals in response to Netrin to regulate the cytoskeleton and also acts independently of Netrin to directly regulate transcription during axon guidance in Drosophila. In other contexts, Dcc acts as a tumor suppressor by directly promoting apoptosis. In this study, we report that Fra is required in the Drosophila female germline for the progression of egg chambers through mid-oogenesis. Loss of Fra in the germline, but not the somatic cells of the ovary, results in the degeneration of egg chambers. Although a failure in nutrient sensing and disruptions in egg chamber polarity can result in degeneration at mid-oogenesis, these factors do not appear to be affected in fra germline mutants. However, similar to the degeneration that occurs in those contexts, the cell death effector Dcp-1 is activated in fra germline mutants. The function of Fra in the female germline is independent of Netrin and requires the transcriptional activation domain of Fra. In contrast to the role of Dcc in promoting cell death, our observations reveal a role for Fra in regulating germline survival by inhibiting apoptosis.
Collapse
Affiliation(s)
| | - Kaitlin M. Laws
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Greg J. Bashaw
- Department of Neuroscience, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
26
|
Girard V, Jollivet F, Knittelfelder O, Celle M, Arsac JN, Chatelain G, Van den Brink DM, Baron T, Shevchenko A, Kühnlein RP, Davoust N, Mollereau B. Abnormal accumulation of lipid droplets in neurons induces the conversion of alpha-Synuclein to proteolytic resistant forms in a Drosophila model of Parkinson's disease. PLoS Genet 2021; 17:e1009921. [PMID: 34788284 PMCID: PMC8635402 DOI: 10.1371/journal.pgen.1009921] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 12/01/2021] [Accepted: 11/02/2021] [Indexed: 11/18/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disorder characterized by alpha-synuclein (αSyn) aggregation and associated with abnormalities in lipid metabolism. The accumulation of lipids in cytoplasmic organelles called lipid droplets (LDs) was observed in cellular models of PD. To investigate the pathophysiological consequences of interactions between αSyn and proteins that regulate the homeostasis of LDs, we used a transgenic Drosophila model of PD, in which human αSyn is specifically expressed in photoreceptor neurons. We first found that overexpression of the LD-coating proteins Perilipin 1 or 2 (dPlin1/2), which limit the access of lipases to LDs, markedly increased triacylglyclerol (TG) loaded LDs in neurons. However, dPlin-induced-LDs in neurons are independent of lipid anabolic (diacylglycerol acyltransferase 1/midway, fatty acid transport protein/dFatp) and catabolic (brummer TG lipase) enzymes, indicating that alternative mechanisms regulate neuronal LD homeostasis. Interestingly, the accumulation of LDs induced by various LD proteins (dPlin1, dPlin2, CG7900 or KlarsichtLD-BD) was synergistically amplified by the co-expression of αSyn, which localized to LDs in both Drosophila photoreceptor neurons and in human neuroblastoma cells. Finally, the accumulation of LDs increased the resistance of αSyn to proteolytic digestion, a characteristic of αSyn aggregation in human neurons. We propose that αSyn cooperates with LD proteins to inhibit lipolysis and that binding of αSyn to LDs contributes to the pathogenic misfolding and aggregation of αSyn in neurons. Parkinson’s disease (PD) is a neurodegenerative disease characterized by the neurotoxic aggregation of the alpha-synuclein (αSyn) protein. Cellular models of the disease are also associated with an abnormal fat storage in the form of lipid droplets (LDs). However, in which cells, neuron or glial cells, LDs accumulate in the organism remains unknown. To understand the relationship between αSyn and the accumulation of LDs, we used a Drosophila (fruit fly) model of PD. We found that, in the presence of a protein that coats LDs, perilipin, LDs accumulate in photoreceptor neurons of the fly. Interestingly, the accumulation of LDs induced by perilipin or other LD-coating proteins was enhanced in the presence of αSyn. Using human neuronal cell lines and the fly, we could show that LD-coating and αSyn proteins localize at the surface of LDs. Finally, we observed that the process of αSyn aggregation was enhanced in the presence of LDs by using a biochemical approach. We thus propose that the association of αSyn with LDs could contribute to αSyn aggregation and progression of the pathology.
Collapse
Affiliation(s)
- Victor Girard
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/ENS de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France
| | - Florence Jollivet
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/ENS de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France
| | - Oskar Knittelfelder
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Marion Celle
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/ENS de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France
| | - Jean-Noel Arsac
- Neurodegenerative Disease Unit; French Agency for Food, Environmental and Occupational Health & Safety Laboratory (Anses) of Lyon, University of Lyon, Lyon, France
| | - Gilles Chatelain
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/ENS de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France
| | - Daan M. Van den Brink
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/ENS de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France
- Plant Systems Physiology, Radboud Institute for Biological and Environmental Sciences, Radboud University, Nijmegen, The Netherlands
| | - Thierry Baron
- Neurodegenerative Disease Unit; French Agency for Food, Environmental and Occupational Health & Safety Laboratory (Anses) of Lyon, University of Lyon, Lyon, France
| | - Andrej Shevchenko
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ronald P. Kühnlein
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
- Field of Excellence BioHealth—University of Graz, Graz, Austria
| | - Nathalie Davoust
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/ENS de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France
- * E-mail: (ND); (BM)
| | - Bertrand Mollereau
- Laboratory of Biology and Modelling of the Cell, UMR5239 CNRS/ENS de Lyon, INSERM U1210, UMS 3444 Biosciences Lyon Gerland, University of Lyon, Lyon, France
- * E-mail: (ND); (BM)
| |
Collapse
|
27
|
Wat LW, Chowdhury ZS, Millington JW, Biswas P, Rideout EJ. Sex determination gene transformer regulates the male-female difference in Drosophila fat storage via the adipokinetic hormone pathway. eLife 2021; 10:e72350. [PMID: 34672260 PMCID: PMC8594944 DOI: 10.7554/elife.72350] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Sex differences in whole-body fat storage exist in many species. For example, Drosophila females store more fat than males. Yet, the mechanisms underlying this sex difference in fat storage remain incompletely understood. Here, we identify a key role for sex determination gene transformer (tra) in regulating the male-female difference in fat storage. Normally, a functional Tra protein is present only in females, where it promotes female sexual development. We show that loss of Tra in females reduced whole-body fat storage, whereas gain of Tra in males augmented fat storage. Tra's role in promoting fat storage was largely due to its function in neurons, specifically the Adipokinetic hormone (Akh)-producing cells (APCs). Our analysis of Akh pathway regulation revealed a male bias in APC activity and Akh pathway function, where this sex-biased regulation influenced the sex difference in fat storage by limiting triglyceride accumulation in males. Importantly, Tra loss in females increased Akh pathway activity, and genetically manipulating the Akh pathway rescued Tra-dependent effects on fat storage. This identifies sex-specific regulation of Akh as one mechanism underlying the male-female difference in whole-body triglyceride levels, and provides important insight into the conserved mechanisms underlying sexual dimorphism in whole-body fat storage.
Collapse
Affiliation(s)
- Lianna W Wat
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Zahid S Chowdhury
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Jason W Millington
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Puja Biswas
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| |
Collapse
|
28
|
Heier C, Klishch S, Stilbytska O, Semaniuk U, Lushchak O. The Drosophila model to interrogate triacylglycerol biology. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158924. [PMID: 33716135 DOI: 10.1016/j.bbalip.2021.158924] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/24/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022]
Abstract
The deposition of storage fat in the form of triacylglycerol (TAG) is an evolutionarily conserved strategy to cope with fluctuations in energy availability and metabolic stress. Organismal TAG storage in specialized adipose tissues provides animals a metabolic reserve that sustains survival during development and starvation. On the other hand, excessive accumulation of adipose TAG, defined as obesity, is associated with an increasing prevalence of human metabolic diseases. During the past decade, the fruit fly Drosophila melanogaster, traditionally used in genetics and developmental biology, has been established as a versatile model system to study TAG metabolism and the etiology of lipid-associated metabolic diseases. Similar to humans, Drosophila TAG homeostasis relies on the interplay of organ systems specialized in lipid uptake, synthesis, and processing, which are integrated by an endocrine network of hormones and messenger molecules. Enzymatic formation of TAG from sugar or dietary lipid, its storage in lipid droplets, and its mobilization by lipolysis occur via mechanisms largely conserved between Drosophila and humans. Notably, dysfunctional Drosophila TAG homeostasis occurs in the context of aging, overnutrition, or defective gene function, and entails tissue-specific and organismal pathologies that resemble human disease. In this review, we summarize the physiology and biochemistry of TAG in Drosophila and outline the potential of this organism as a model system to understand the genetic and dietary basis of TAG storage and TAG-related metabolic disorders.
Collapse
Affiliation(s)
- Christoph Heier
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Humboldtstrasse 50, A-8010 Graz, Austria; BioTechMed-Graz, Graz, Austria.
| | - Svitlana Klishch
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Olha Stilbytska
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Uliana Semaniuk
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
29
|
Clerbaux LA, Schultz H, Roman-Holba S, Ruan DF, Yu R, Lamb AM, Bommer GT, Kennell JA. The microRNA miR-33 is a pleiotropic regulator of metabolic and developmental processes in Drosophila melanogaster. Dev Dyn 2021; 250:1634-1650. [PMID: 33840153 DOI: 10.1002/dvdy.344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/31/2021] [Accepted: 04/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND miR-33 family members are well characterized regulators of cellular lipid levels in mammals. Previous studies have shown that overexpression of miR-33 in Drosophila melanogaster leads to elevated triacylglycerol (TAG) levels in certain contexts. Although loss of miR-33 in flies causes subtle defects in larval and adult ovaries, the effects of miR-33 deficiency on lipid metabolism and other phenotypes impacted by metabolic state have not yet been characterized. RESULTS We found that loss of miR-33 predisposes flies to elevated TAG levels, and we identified genes involved in TAG synthesis as direct targets of miR-33, including atpcl, midway, and Akt1. miR-33 mutants survived longer upon starvation but showed greater sensitivity to an oxidative stressor. We also found evidence that miR-33 is a negative regulator of cuticle pigmentation and that miR-33 mutants show a reduction in interfollicular stalk cells during oogenesis. CONCLUSION Our data suggest that miR-33 is a conserved regulator of lipid homeostasis, and its targets are involved in both degradation and synthesis of fatty acids and TAG. The constellation of phenotypes involving tissues that are highly sensitive to metabolic state suggests that miR-33 serves to prevent extreme fluctuations in metabolically sensitive tissues.
Collapse
Affiliation(s)
- Laure-Alix Clerbaux
- Laboratory of Physiological Chemistry, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium.,Department of Biology and Program in Biochemistry, Vassar College, Poughkeepsie, New York, USA
| | - Hayley Schultz
- Department of Biology and Program in Biochemistry, Vassar College, Poughkeepsie, New York, USA
| | - Samara Roman-Holba
- Department of Biology and Program in Biochemistry, Vassar College, Poughkeepsie, New York, USA
| | - Dan Fu Ruan
- Department of Biology and Program in Biochemistry, Vassar College, Poughkeepsie, New York, USA
| | - Ronald Yu
- Department of Biology and Program in Biochemistry, Vassar College, Poughkeepsie, New York, USA
| | - Abigail M Lamb
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Guido T Bommer
- Laboratory of Physiological Chemistry, de Duve Institute, Université Catholique de Louvain, Bruxelles, Belgium
| | - Jennifer A Kennell
- Department of Biology and Program in Biochemistry, Vassar College, Poughkeepsie, New York, USA
| |
Collapse
|
30
|
Blumrich A, Vogler G, Dresen S, Diop SB, Jaeger C, Leberer S, Grune J, Wirth EK, Hoeft B, Renko K, Foryst-Ludwig A, Spranger J, Sigrist S, Bodmer R, Kintscher U. Fat-body brummer lipase determines survival and cardiac function during starvation in Drosophila melanogaster. iScience 2021; 24:102288. [PMID: 33889813 PMCID: PMC8050372 DOI: 10.1016/j.isci.2021.102288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 12/17/2022] Open
Abstract
The cross talk between adipose tissue and the heart has an increasing importance for cardiac function under physiological and pathological conditions. This study characterizes the role of fat body lipolysis for cardiac function in Drosophila melanogaster. Perturbation of the function of the key lipolytic enzyme, brummer (bmm), an ortholog of the mammalian ATGL (adipose triglyceride lipase) exclusively in the fly's fat body, protected the heart against starvation-induced dysfunction. We further provide evidence that this protection is caused by the preservation of glycerolipid stores, resulting in a starvation-resistant maintenance of energy supply and adequate cardiac ATP synthesis. Finally, we suggest that alterations of lipolysis are tightly coupled to lipogenic processes, participating in the preservation of lipid energy substrates during starvation. Thus, we identified the inhibition of adipose tissue lipolysis and subsequent energy preservation as a protective mechanism against cardiac dysfunction during catabolic stress. A cross talk between fat body and the heart regulates cardiac function in Drosophila Knockdown of fat-body brummer lipase prevents starvation-induced cardiac dysfunction This involves preservation of lipid stores and maintenance of cardiac energy supply Brummer-mediated preservation of fat body lipid stores involves lipolysis and lipogenesis
Collapse
Affiliation(s)
- Annelie Blumrich
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, CCR, Hessische Str. 3-4, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Georg Vogler
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sandra Dresen
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, CCR, Hessische Str. 3-4, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Soda Balla Diop
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Carsten Jaeger
- Federal Institute for Materials Research and Testing (BAM), Berlin, Germany
| | - Sarah Leberer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, CCR, Hessische Str. 3-4, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Jana Grune
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Physiology, Berlin, Germany
| | - Eva K. Wirth
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, Berlin, Germany
| | - Beata Hoeft
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, CCR, Hessische Str. 3-4, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Kostja Renko
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Experimental Endocrinology, Berlin, Germany
| | - Anna Foryst-Ludwig
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, CCR, Hessische Str. 3-4, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Joachim Spranger
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology and Metabolism, Berlin, Germany
| | - Stephan Sigrist
- Institute of Biology/Genetics, Freie Universität Berlin, Berlin, Germany
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Ulrich Kintscher
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pharmacology, Center for Cardiovascular Research, CCR, Hessische Str. 3-4, 10115 Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Corresponding author
| |
Collapse
|
31
|
Toprak U, Hegedus D, Doğan C, Güney G. A journey into the world of insect lipid metabolism. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2020; 104:e21682. [PMID: 32335968 DOI: 10.1002/arch.21682] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/08/2020] [Accepted: 04/08/2020] [Indexed: 06/11/2023]
Abstract
Lipid metabolism is fundamental to life. In insects, it is critical, during reproduction, flight, starvation, and diapause. The coordination center for insect lipid metabolism is the fat body, which is analogous to the vertebrate adipose tissue and liver. Fat body contains various different cell types; however, adipocytes and oenocytes are the primary cells related to lipid metabolism. Lipid metabolism starts with the hydrolysis of dietary lipids, absorption of lipid monomers, followed by lipid transport from midgut to the fat body, lipogenesis or lipolysis in the fat body, and lipid transport from fat body to other sites demanding energy. Lipid metabolism is under the control of hormones, transcription factors, secondary messengers and posttranscriptional modifications. Primarily, lipogenesis is under the control of insulin-like peptides that activate lipogenic transcription factors, such as sterol regulatory element-binding proteins, whereas lipolysis is coordinated by the adipokinetic hormone that activates lipolytic transcription factors, such as forkhead box class O and cAMP-response element-binding protein. Calcium is the primary-secondary messenger affecting lipid metabolism and has different outcomes depending on the site of lipogenesis or lipolysis. Phosphorylation is central to lipid metabolism and multiple phosphorylases are involved in lipid accumulation or hydrolysis. Although most of the knowledge of insect lipid metabolism comes from the studies on the model Drosophila; other insects, in particular those with obligatory or facultative diapause, also have great potential to study lipid metabolism. The use of these models would significantly improve our knowledge of insect lipid metabolism.
Collapse
Affiliation(s)
- Umut Toprak
- Molecular Entomology Laboratory, Department of Plant Protection, Faculty of Agriculture, Ankara University, Ankara, Turkey
| | - Dwayne Hegedus
- Agriculture and Agri-Food Canada, Saskatoon Research Centre, Saskatoon, Saskatchewan, Canada
- Department of Food and Bioproduct Sciences, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Cansu Doğan
- Molecular Entomology Laboratory, Department of Plant Protection, Faculty of Agriculture, Ankara University, Ankara, Turkey
| | - Gözde Güney
- Molecular Entomology Laboratory, Department of Plant Protection, Faculty of Agriculture, Ankara University, Ankara, Turkey
| |
Collapse
|
32
|
Toprak U. The Role of Peptide Hormones in Insect Lipid Metabolism. Front Physiol 2020; 11:434. [PMID: 32457651 PMCID: PMC7221030 DOI: 10.3389/fphys.2020.00434] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Lipids are the primary storage molecules and an essential source of energy in insects during reproduction, prolonged periods of flight, starvation, and diapause. The coordination center for insect lipid metabolism is the fat body, which is analogous to the vertebrate adipose tissue and liver. The fat body is primarily composed of adipocytes, which accumulate triacylglycerols in intracellular lipid droplets. Genomics and proteomics, together with functional analyses, such as RNA interference and CRISPR/Cas9-targeted genome editing, identified various genes involved in lipid metabolism and elucidated their functions. However, the endocrine control of insect lipid metabolism, in particular the roles of peptide hormones in lipogenesis and lipolysis are relatively less-known topics. In the current review, the neuropeptides that directly or indirectly affect insect lipid metabolism are introduced. The primary lipolytic and lipogenic peptide hormones are adipokinetic hormone and the brain insulin-like peptides (ILP2, ILP3, ILP5). Other neuropeptides, such as insulin-growth factor ILP6, neuropeptide F, allatostatin-A, corazonin, leucokinin, tachykinins and limostatin, might stimulate lipolysis, while diapause hormone-pheromone biosynthesis activating neuropeptide, short neuropeptide F, CCHamide-2, and the cytokines Unpaired 1 and Unpaired 2 might induce lipogenesis. Most of these peptides interact with one another, but mostly with insulin signaling, and therefore affect lipid metabolism indirectly. Peptide hormones are also involved in lipid metabolism during reproduction, flight, diapause, starvation, infections and immunity; these are also highlighted. The review concludes with a discussion of the potential of lipid metabolism-related peptide hormones in pest management.
Collapse
Affiliation(s)
- Umut Toprak
- Molecular Entomology Lab., Department of Plant Protection Ankara, Faculty of Agriculture, Ankara University, Ankara, Turkey
| |
Collapse
|
33
|
Wat LW, Chao C, Bartlett R, Buchanan JL, Millington JW, Chih HJ, Chowdhury ZS, Biswas P, Huang V, Shin LJ, Wang LC, Gauthier MPL, Barone MC, Montooth KL, Welte MA, Rideout EJ. A role for triglyceride lipase brummer in the regulation of sex differences in Drosophila fat storage and breakdown. PLoS Biol 2020; 18:e3000595. [PMID: 31961851 PMCID: PMC6994176 DOI: 10.1371/journal.pbio.3000595] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 01/31/2020] [Accepted: 01/03/2020] [Indexed: 01/26/2023] Open
Abstract
Triglycerides are the major form of stored fat in all animals. One important determinant of whole-body fat storage is whether an animal is male or female. Here, we use Drosophila, an established model for studies on triglyceride metabolism, to gain insight into the genes and physiological mechanisms that contribute to sex differences in fat storage. Our analysis of triglyceride storage and breakdown in both sexes identified a role for triglyceride lipase brummer (bmm) in the regulation of sex differences in triglyceride homeostasis. Normally, male flies have higher levels of bmm mRNA both under normal culture conditions and in response to starvation, a lipolytic stimulus. We find that loss of bmm largely eliminates the sex difference in triglyceride storage and abolishes the sex difference in triglyceride breakdown via strongly male-biased effects. Although we show that bmm function in the fat body affects whole-body triglyceride levels in both sexes, in males, we identify an additional role for bmm function in the somatic cells of the gonad and in neurons in the regulation of whole-body triglyceride homeostasis. Furthermore, we demonstrate that lipid droplets are normally present in both the somatic cells of the male gonad and in neurons, revealing a previously unrecognized role for bmm function, and possibly lipid droplets, in these cell types in the regulation of whole-body triglyceride homeostasis. Taken together, our data reveal a role for bmm function in the somatic cells of the gonad and in neurons in the regulation of male–female differences in fat storage and breakdown and identify bmm as a link between the regulation of triglyceride homeostasis and biological sex. An investigation of the genetic and physiological mechanisms underlying sex differences in fat storage and breakdown in the fruit fly Drosophila identifies previously unrecognized sex- and cell type-specific roles for the conserved triglyceride lipase brummer.
Collapse
Affiliation(s)
- Lianna W. Wat
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Charlotte Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Rachael Bartlett
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Justin L. Buchanan
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Jason W. Millington
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Hui Ju Chih
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Zahid S. Chowdhury
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Puja Biswas
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Vivian Huang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Leah J. Shin
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Lin Chuan Wang
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Marie-Pierre L. Gauthier
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Maria C. Barone
- Department of Biology, University of Rochester, Rochester, New York, United States of America
| | - Kristi L. Montooth
- School of Biological Sciences, University of Nebraska-Lincoln, Lincoln, Nebraska, United States of America
| | - Michael A. Welte
- Department of Biology, University of Rochester, Rochester, New York, United States of America
| | - Elizabeth J. Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail:
| |
Collapse
|
34
|
Yao Y, Li X, Wang W, Liu Z, Chen J, Ding M, Huang X. MRT, Functioning with NURF Complex, Regulates Lipid Droplet Size. Cell Rep 2019; 24:2972-2984. [PMID: 30208321 DOI: 10.1016/j.celrep.2018.08.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/08/2018] [Accepted: 08/09/2018] [Indexed: 12/27/2022] Open
Abstract
Lipid droplets (LDs) are highly dynamic organelles that store neutral lipids. Through a gene overexpression screen in the Drosophila larval fat body, we have identified that MRT, an Myb/switching-defective protein 3 (Swi3), Adaptor 2 (Ada2), Nuclear receptor co-repressor (N-CoR), Transcription factor (TF)IIIB (SANT)-like DNA-binding domain-containing protein, regulates LD size and lipid storage. MRT directly interacts with, and is functionally dependent on, the PZG and NURF chromatin-remodeling complex components. MRT binds to the promoter of plin1, the gene encoding the LD-resident protein perilipin, and inhibits the transcription of plin1. In vitro LD coalescence assays suggest that mrt overexpression or loss of plin1 function facilitates LD coalescence. Our findings suggest that MRT functions together with chromatin-remodeling factors to regulate LD size, likely through the transcriptional repression of plin1.
Collapse
Affiliation(s)
- Yan Yao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xia Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wei Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhonghua Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jianming Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Mei Ding
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xun Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
35
|
Harsh S, Heryanto C, Eleftherianos I. Intestinal lipid droplets as novel mediators of host-pathogen interaction in Drosophila. Biol Open 2019; 8:bio.039040. [PMID: 31278163 PMCID: PMC6679391 DOI: 10.1242/bio.039040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Lipid droplets (LDs) are lipid-carrying multifunctional organelles, which might also interact with pathogens and influence the host immune response. However, the exact nature of these interactions remains currently unexplored. Here we show that systemic infection of Drosophila adult flies with non-pathogenic Escherichia coli, the extracellular bacterial pathogen Photorhabdus luminescens or the facultative intracellular pathogen Photorhabdus asymbiotica results in intestinal steatosis marked by lipid accumulation in the midgut. Accumulation of LDs in the midgut also correlates with increased whole-body lipid levels characterized by increased expression of genes regulating lipogenesis. The lipid-enriched midgut further displays reduced expression of the enteroendocrine-secreted hormone, Tachykinin. The observed lipid accumulation requires the Gram-negative cell wall pattern recognition molecule, PGRP-LC, but not PGRP-LE, for the humoral immune response. Altogether, our findings indicate that Drosophila LDs are inducible organelles, which can serve as markers for inflammation and, depending on the nature of the challenge, they can dictate the outcome of the infection. Summary: Lipid droplets are inducible organelles, act as inflammatory markers and, depending on the nature of challenge, can dictate the outcome of the infection.
Collapse
Affiliation(s)
- Sneh Harsh
- Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington DC 20052, USA
| | - Christa Heryanto
- Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington DC 20052, USA
| | - Ioannis Eleftherianos
- Department of Biological Sciences, Institute for Biomedical Sciences, The George Washington University, Washington DC 20052, USA
| |
Collapse
|
36
|
Hehlert P, Hofferek V, Heier C, Eichmann TO, Riedel D, Rosenberg J, Takaćs A, Nagy HM, Oberer M, Zimmermann R, Kühnlein RP. The α/β-hydrolase domain-containing 4- and 5-related phospholipase Pummelig controls energy storage in Drosophila. J Lipid Res 2019; 60:1365-1378. [PMID: 31164391 DOI: 10.1194/jlr.m092817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 06/03/2019] [Indexed: 01/05/2023] Open
Abstract
Triglycerides (TGs) are the main energy storage form that accommodates changing organismal energy demands. In Drosophila melanogaster, the TG lipase Brummer is centrally important for body fat mobilization. Its gene brummer (bmm) encodes the ortholog of mammalian adipose TG lipase, which becomes activated by α/β-hydrolase domain-containing 5 (ABHD5/CGI-58), one member of the paralogous gene pair, α/β-hydrolase domain-containing 4 (ABHD4) and ABHD5 In Drosophila, the pummelig (puml) gene encodes the single sequence-related protein to mammalian ABHD4/ABHD5 with unknown function. We generated puml deletion mutant flies, that were short-lived as a result of lipid metabolism changes, stored excess body fat at the expense of glycogen, and exhibited ectopic fat storage with altered TG FA profile in the fly kidneys, called Malpighian tubules. TG accumulation in puml mutants was not associated with increased food intake but with elevated lipogenesis; starvation-induced lipid mobilization remained functional. Despite its structural similarity to mammalian ABHD5, Puml did not stimulate TG lipase activity of Bmm in vitro. Rather, Puml acted as a phospholipase that localized on lipid droplets, mitochondria, and peroxisomes. Together, these results show that the ABHD4/5 family member Puml is a versatile phospholipase that regulates Drosophila body fat storage and energy metabolism.
Collapse
Affiliation(s)
- Philip Hehlert
- Research Group Molecular Physiology Max-Planck-Institut für Biophysikalische Chemie, Göttingen, Germany
| | - Vinzenz Hofferek
- Max-Planck-Institut für Molekulare Pflanzenphysiologie Potsdam, Germany
| | - Christoph Heier
- Institute of Molecular Biosciences University of Graz, Graz, Austria
| | - Thomas O Eichmann
- Institute of Molecular Biosciences University of Graz, Graz, Austria
| | - Dietmar Riedel
- Department of Structural Dynamics, Electron Microscopy, Max-Planck-Institut für Biophysikalische Chemie, Göttingen, Germany
| | - Jonathan Rosenberg
- Research Group Molecular Physiology Max-Planck-Institut für Biophysikalische Chemie, Göttingen, Germany
| | - Anna Takaćs
- Research Group Molecular Physiology Max-Planck-Institut für Biophysikalische Chemie, Göttingen, Germany
| | - Harald M Nagy
- Institute of Molecular Biosciences University of Graz, Graz, Austria
| | - Monika Oberer
- Institute of Molecular Biosciences University of Graz, Graz, Austria.,BioTechMed-Graz Graz, Austria
| | - Robert Zimmermann
- Institute of Molecular Biosciences University of Graz, Graz, Austria.,BioTechMed-Graz Graz, Austria
| | - Ronald P Kühnlein
- Research Group Molecular Physiology Max-Planck-Institut für Biophysikalische Chemie, Göttingen, Germany .,Institute of Molecular Biosciences University of Graz, Graz, Austria.,BioTechMed-Graz Graz, Austria
| |
Collapse
|
37
|
Wu Z, Liu JL. Cytoophidia respond to nutrient stress in Drosophila. Exp Cell Res 2019; 376:159-167. [PMID: 30768932 PMCID: PMC6403103 DOI: 10.1016/j.yexcr.2019.02.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Revised: 01/13/2019] [Accepted: 02/09/2019] [Indexed: 01/08/2023]
Abstract
CTP synthase (CTPsyn) is a metabolic enzyme essential for the de novo synthesis of CTP the nucleotide. CTPsyn can be compartmented into filamentous structures named cytoophidia. Cytoophidia are conserved in a wide range of species and are highly abundant in Drosophila ovaries. Here we report that cytoophidia elongate upon nutrient deprivation, CTPsyn overexpression or heat shock in Drosophila ovaries. We also show that the curvature of cytoophidia changes during apoptosis. Moreover, cytoophidia can be transported from nurse cells to the oocyte via ring canals. Our study demonstrates that cytoophidia can respond to stress and are very dynamic in Drosophila ovaries.
Collapse
Affiliation(s)
- Zheng Wu
- School of Life Science and Technology, ShanghaiTech University, 230 Haike Road, 201210 Shanghai, China
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech University, 230 Haike Road, 201210 Shanghai, China; MRC Functional Genomics Unit, Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3PT, United Kingdom.
| |
Collapse
|
38
|
Triacylglycerol Metabolism in Drosophila melanogaster. Genetics 2019; 210:1163-1184. [PMID: 30523167 DOI: 10.1534/genetics.118.301583] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022] Open
Abstract
Triacylglycerol (TAG) is the most important caloric source with respect to energy homeostasis in animals. In addition to its evolutionarily conserved importance as an energy source, TAG turnover is crucial to the metabolism of structural and signaling lipids. These neutral lipids are also key players in development and disease. Here, we review the metabolism of TAG in the Drosophila model system. Recently, the fruit fly has attracted renewed attention in research due to the unique experimental approaches it affords in studying the tissue-autonomous and interorgan regulation of lipid metabolism in vivo Following an overview of the systemic control of fly body fat stores, we will cover lipid anabolic, enzymatic, and regulatory processes, which begin with the dietary lipid breakdown and de novo lipogenesis that results in lipid droplet storage. Next, we focus on lipolytic processes, which mobilize storage TAG to make it metabolically accessible as either an energy source or as a building block for biosynthesis of other lipid classes. Since the buildup and breakdown of fat involves various organs, we highlight avenues of lipid transport, which are at the heart of functional integration of organismic lipid metabolism. Finally, we draw attention to some "missing links" in basic neutral lipid metabolism and conclude with a perspective on how fly research can be exploited to study functional metabolic roles of diverse lipids.
Collapse
|
39
|
Effect of Larval Nutrition on Maternal mRNA Contribution to the Drosophila Egg. G3-GENES GENOMES GENETICS 2018; 8:1933-1941. [PMID: 29666195 PMCID: PMC5982822 DOI: 10.1534/g3.118.200283] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Embryonic development begins under the control of maternal gene products, mRNAs and proteins that the mother deposits into the egg; the zygotic genome is activated some time later. Maternal control of early development is conserved across metazoans. Gene products contributed by mothers are critical to many early developmental processes, and set up trajectories for the rest of development. Maternal deposition of these factors is an often-overlooked aspect of parental investment. If the mother experiences challenging environmental conditions, such as poor nutrition, previous studies in Drosophila melanogaster have demonstrated a plastic response wherein these mothers may produce larger eggs to buffer the offspring against the same difficult environment. This additional investment can produce offspring that are more fit in the challenging environment. With this study, we ask whether D. melanogaster mothers who experience poor nutrition during their own development change their gene product contribution to the egg. We perform mRNA-Seq on eggs at a stage where all mRNAs are maternally derived, from mothers with different degrees of nutritional limitation. We find that nutritional limitation produces similar transcript changes at all degrees of limitation tested. Genes that have lower transcript abundance in nutritionally limited mothers are those involved in translation, which is likely one of the most energetically costly processes occurring in the early embryo. We find an increase in transcripts for transport and localization of macromolecules, and for the electron transport chain. The eggs produced by nutrition-limited mothers show a plastic response in mRNA deposition, which may better prepare the future embryo for development in a nutrition-limited environment.
Collapse
|
40
|
Abstract
Excess adipose fat accumulation, or obesity, is a growing problem worldwide in terms of both the rate of incidence and the severity of obesity-associated metabolic disease. Adipose tissue evolved in animals as a specialized dynamic lipid storage depot: adipose cells synthesize fat (a process called lipogenesis) when energy is plentiful and mobilize stored fat (a process called lipolysis) when energy is needed. When a disruption of lipid homeostasis favors increased fat synthesis and storage with little turnover owing to genetic predisposition, overnutrition or sedentary living, complications such as diabetes and cardiovascular disease are more likely to arise. The vinegar fly Drosophila melanogaster (Diptera: Drosophilidae) is used as a model to better understand the mechanisms governing fat metabolism and distribution. Flies offer a wealth of paradigms with which to study the regulation and physiological effects of fat accumulation. Obese flies accumulate triacylglycerols in the fat body, an organ similar to mammalian adipose tissue, which specializes in lipid storage and catabolism. Discoveries in Drosophila have ranged from endocrine hormones that control obesity to subcellular mechanisms that regulate lipogenesis and lipolysis, many of which are evolutionarily conserved. Furthermore, obese flies exhibit pathophysiological complications, including hyperglycemia, reduced longevity and cardiovascular function - similar to those observed in obese humans. Here, we review some of the salient features of the fly that enable researchers to study the contributions of feeding, absorption, distribution and the metabolism of lipids to systemic physiology.
Collapse
Affiliation(s)
- Laura Palanker Musselman
- Department of Biological Sciences, Binghamton University, State University of New York, Binghamton, NY 13902, USA
| | - Ronald P Kühnlein
- Department of Biochemistry 1, Institute of Molecular Biosciences, University of Graz, Humboldtstraβe 50/II, A-8010 Graz, Austria.,BioTechMed-Graz, Graz, Austria
| |
Collapse
|
41
|
Lehmann M. Endocrine and physiological regulation of neutral fat storage in Drosophila. Mol Cell Endocrinol 2018; 461:165-177. [PMID: 28893568 PMCID: PMC5756521 DOI: 10.1016/j.mce.2017.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/06/2017] [Accepted: 09/08/2017] [Indexed: 12/20/2022]
Abstract
After having revolutionized our understanding of the mechanisms of animal development, Drosophila melanogaster has more recently emerged as an equally valid genetic model in the field of animal metabolism. An increasing number of studies have revealed that many signaling pathways that control metabolism in mammals, including pathways controlled by nutrients (insulin, TOR), steroid hormone, glucagon, and hedgehog, are functionally conserved between mammals and Drosophila. In fact, genetic screens and analyses in Drosophila have identified new players and filled in gaps in the signaling networks that control metabolism. This review focuses on data that show how these networks control the formation and breakdown of triacylglycerol energy stores in the fat tissue of Drosophila.
Collapse
Affiliation(s)
- Michael Lehmann
- Department of Biological Sciences, SCEN 601, 1 University of Arkansas, Fayetteville, AR 72701, USA.
| |
Collapse
|
42
|
McMillan EA, Longo SM, Smith MD, Broskin S, Lin B, Singh NK, Strochlic TI. The protein kinase CK2 substrate Jabba modulates lipid metabolism during Drosophila oogenesis. J Biol Chem 2018; 293:2990-3002. [PMID: 29326167 DOI: 10.1074/jbc.m117.814657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/07/2018] [Indexed: 11/06/2022] Open
Abstract
Lipid metabolism plays a critical role in female reproduction. During oogenesis, maturing oocytes accumulate high levels of neutral lipids that are essential for both energy production and the synthesis of other lipid molecules. Metabolic pathways within the ovary are partially regulated by protein kinases that link metabolic status to oocyte development. Although the functions of several kinases in this process are well established, the roles that many other kinases play in coordinating metabolic state with female germ cell development are unknown. Here, we demonstrate that the catalytic activity of casein kinase 2 (CK2) is essential for Drosophila oogenesis. Using an unbiased biochemical screen that leveraged an unusual catalytic property of the kinase, we identified a novel CK2 substrate in the Drosophila ovary, the lipid droplet-associated protein Jabba. We show that Jabba is essential for modulating ovarian lipid metabolism and for regulating female fertility in the fly. Our findings shed light on a CK2-dependent signaling pathway governing lipid metabolism in the ovary and provide insight into the long-recognized but poorly understood association between energy metabolism and female reproduction.
Collapse
Affiliation(s)
- Emily A McMillan
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Sheila M Longo
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Michael D Smith
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Sarah Broskin
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Baicheng Lin
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Nisha K Singh
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102
| | - Todd I Strochlic
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania 19102.
| |
Collapse
|
43
|
Sieber MH, Spradling AC. The role of metabolic states in development and disease. Curr Opin Genet Dev 2017; 45:58-68. [PMID: 28347941 PMCID: PMC6894399 DOI: 10.1016/j.gde.2017.03.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/23/2017] [Accepted: 03/02/2017] [Indexed: 12/17/2022]
Abstract
During development, cells adopt distinct metabolic strategies to support growth, produce energy, and meet the demands of a mature tissue. Some of these metabolic states maintain a constrained program of nutrient utilization, while others providing metabolic flexibility as a means to couple developmental progression with nutrient availability. Here we discuss our understanding of metabolic programs, and how they support specific aspects of animal development. During phases of rapid proliferation a subset of metabolic programs provide the building blocks to support growth. During differentiation, metabolic programs shift to support the unique demands of each tissue. Finally, we discuss how a model system, such as Drosophila egg development, can provide a versatile platform to discover novel mechanisms controlling programmed shift in metabolism.
Collapse
Affiliation(s)
- Matthew H Sieber
- Department of Embryology, Howard Hughes Medical Institute Labs, Carnegie Institution for Science, Baltimore, MD 21218, United States
| | - Allan C Spradling
- Department of Embryology, Howard Hughes Medical Institute Labs, Carnegie Institution for Science, Baltimore, MD 21218, United States.
| |
Collapse
|
44
|
Regulation of phagocyte triglyceride by a STAT-ATG2 pathway controls mycobacterial infection. Nat Commun 2017; 8:14642. [PMID: 28262681 PMCID: PMC5343520 DOI: 10.1038/ncomms14642] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/18/2017] [Indexed: 01/27/2023] Open
Abstract
Mycobacterium tuberculosis remains a global threat to human health, yet the molecular mechanisms regulating immunity remain poorly understood. Cytokines can promote or inhibit mycobacterial survival inside macrophages and the underlying mechanisms represent potential targets for host-directed therapies. Here we show that cytokine-STAT signalling promotes mycobacterial survival within macrophages by deregulating lipid droplets via ATG2 repression. In Drosophila infected with Mycobacterium marinum, mycobacterium-induced STAT activity triggered by unpaired-family cytokines reduces Atg2 expression, permitting deregulation of lipid droplets. Increased Atg2 expression or reduced macrophage triglyceride biosynthesis, normalizes lipid deposition in infected phagocytes and reduces numbers of viable intracellular mycobacteria. In human macrophages, addition of IL-6 promotes mycobacterial survival and BCG-induced lipid accumulation by a similar, but probably not identical, mechanism. Our results reveal Atg2 regulation as a mechanism by which cytokines can control lipid droplet homeostasis and consequently resistance to mycobacterial infection in Drosophila. Cytokines and their associated pathways can affect survival of Mycobacterium tuberculosis in macrophages, representing potential targets for host-directed therapies. Here, Péan et al. show that cytokine-STAT signalling promotes mycobacterial survival within macrophages by deregulating lipid droplet homeostasis.
Collapse
|
45
|
Kanost MR, Arrese EL, Cao X, Chen YR, Chellapilla S, Goldsmith MR, Grosse-Wilde E, Heckel DG, Herndon N, Jiang H, Papanicolaou A, Qu J, Soulages JL, Vogel H, Walters J, Waterhouse RM, Ahn SJ, Almeida FC, An C, Aqrawi P, Bretschneider A, Bryant WB, Bucks S, Chao H, Chevignon G, Christen JM, Clarke DF, Dittmer NT, Ferguson LCF, Garavelou S, Gordon KHJ, Gunaratna RT, Han Y, Hauser F, He Y, Heidel-Fischer H, Hirsh A, Hu Y, Jiang H, Kalra D, Klinner C, König C, Kovar C, Kroll AR, Kuwar SS, Lee SL, Lehman R, Li K, Li Z, Liang H, Lovelace S, Lu Z, Mansfield JH, McCulloch KJ, Mathew T, Morton B, Muzny DM, Neunemann D, Ongeri F, Pauchet Y, Pu LL, Pyrousis I, Rao XJ, Redding A, Roesel C, Sanchez-Gracia A, Schaack S, Shukla A, Tetreau G, Wang Y, Xiong GH, Traut W, Walsh TK, Worley KC, Wu D, Wu W, Wu YQ, Zhang X, Zou Z, Zucker H, Briscoe AD, Burmester T, Clem RJ, Feyereisen R, Grimmelikhuijzen CJP, Hamodrakas SJ, Hansson BS, Huguet E, Jermiin LS, Lan Q, Lehman HK, Lorenzen M, Merzendorfer H, Michalopoulos I, Morton DB, Muthukrishnan S, Oakeshott JG, Palmer W, Park Y, Passarelli AL, et alKanost MR, Arrese EL, Cao X, Chen YR, Chellapilla S, Goldsmith MR, Grosse-Wilde E, Heckel DG, Herndon N, Jiang H, Papanicolaou A, Qu J, Soulages JL, Vogel H, Walters J, Waterhouse RM, Ahn SJ, Almeida FC, An C, Aqrawi P, Bretschneider A, Bryant WB, Bucks S, Chao H, Chevignon G, Christen JM, Clarke DF, Dittmer NT, Ferguson LCF, Garavelou S, Gordon KHJ, Gunaratna RT, Han Y, Hauser F, He Y, Heidel-Fischer H, Hirsh A, Hu Y, Jiang H, Kalra D, Klinner C, König C, Kovar C, Kroll AR, Kuwar SS, Lee SL, Lehman R, Li K, Li Z, Liang H, Lovelace S, Lu Z, Mansfield JH, McCulloch KJ, Mathew T, Morton B, Muzny DM, Neunemann D, Ongeri F, Pauchet Y, Pu LL, Pyrousis I, Rao XJ, Redding A, Roesel C, Sanchez-Gracia A, Schaack S, Shukla A, Tetreau G, Wang Y, Xiong GH, Traut W, Walsh TK, Worley KC, Wu D, Wu W, Wu YQ, Zhang X, Zou Z, Zucker H, Briscoe AD, Burmester T, Clem RJ, Feyereisen R, Grimmelikhuijzen CJP, Hamodrakas SJ, Hansson BS, Huguet E, Jermiin LS, Lan Q, Lehman HK, Lorenzen M, Merzendorfer H, Michalopoulos I, Morton DB, Muthukrishnan S, Oakeshott JG, Palmer W, Park Y, Passarelli AL, Rozas J, Schwartz LM, Smith W, Southgate A, Vilcinskas A, Vogt R, Wang P, Werren J, Yu XQ, Zhou JJ, Brown SJ, Scherer SE, Richards S, Blissard GW. Multifaceted biological insights from a draft genome sequence of the tobacco hornworm moth, Manduca sexta. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 76:118-147. [PMID: 27522922 PMCID: PMC5010457 DOI: 10.1016/j.ibmb.2016.07.005] [Show More Authors] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 06/27/2016] [Accepted: 07/14/2016] [Indexed: 05/19/2023]
Abstract
Manduca sexta, known as the tobacco hornworm or Carolina sphinx moth, is a lepidopteran insect that is used extensively as a model system for research in insect biochemistry, physiology, neurobiology, development, and immunity. One important benefit of this species as an experimental model is its extremely large size, reaching more than 10 g in the larval stage. M. sexta larvae feed on solanaceous plants and thus must tolerate a substantial challenge from plant allelochemicals, including nicotine. We report the sequence and annotation of the M. sexta genome, and a survey of gene expression in various tissues and developmental stages. The Msex_1.0 genome assembly resulted in a total genome size of 419.4 Mbp. Repetitive sequences accounted for 25.8% of the assembled genome. The official gene set is comprised of 15,451 protein-coding genes, of which 2498 were manually curated. Extensive RNA-seq data from many tissues and developmental stages were used to improve gene models and for insights into gene expression patterns. Genome wide synteny analysis indicated a high level of macrosynteny in the Lepidoptera. Annotation and analyses were carried out for gene families involved in a wide spectrum of biological processes, including apoptosis, vacuole sorting, growth and development, structures of exoskeleton, egg shells, and muscle, vision, chemosensation, ion channels, signal transduction, neuropeptide signaling, neurotransmitter synthesis and transport, nicotine tolerance, lipid metabolism, and immunity. This genome sequence, annotation, and analysis provide an important new resource from a well-studied model insect species and will facilitate further biochemical and mechanistic experimental studies of many biological systems in insects.
Collapse
Affiliation(s)
- Michael R Kanost
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA.
| | - Estela L Arrese
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Xiaolong Cao
- Department of Entomology and Plant Pathology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Yun-Ru Chen
- Boyce Thompson Institute at Cornell University, Tower Road, Ithaca, NY, 14853, USA
| | - Sanjay Chellapilla
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Marian R Goldsmith
- Biological Sciences Department, University of Rhode Island, Kingston, RI, 02881, USA
| | - Ewald Grosse-Wilde
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knoell-Strasse, 8, D-07745, Jena, Germany
| | - David G Heckel
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - Nicolae Herndon
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Haobo Jiang
- Department of Entomology and Plant Pathology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Alexie Papanicolaou
- Hawkesbury Institute for the Environment, Western Sydney University, Richmond, NSW, 2753, Australia
| | - Jiaxin Qu
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Jose L Soulages
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Heiko Vogel
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - James Walters
- Department of Ecology and Evolutionary Biology, Univ. Kansas, Lawrence, KS, 66045, USA
| | - Robert M Waterhouse
- Department of Genetic Medicine and Development, University of Geneva Medical School, rue Michel-Servet 1, 1211, Geneva, Switzerland; Swiss Institute of Bioinformatics, rue Michel-Servet 1, 1211, Geneva, Switzerland; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA, 02139, USA; The Broad Institute of MIT and Harvard, Cambridge, 415 Main Street, MA, 02142, USA
| | - Seung-Joon Ahn
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - Francisca C Almeida
- Departament de Genètica and Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Barcelona, Spain
| | - Chunju An
- Department of Entomology, China Agricultural University, Beijing, China
| | - Peshtewani Aqrawi
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Anne Bretschneider
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - William B Bryant
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Sascha Bucks
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knoell-Strasse, 8, D-07745, Jena, Germany
| | - Hsu Chao
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Germain Chevignon
- Institut de Recherche sur la Biologie de l'Insecte, UMR CNRS 7261, UFR Sciences et Techniques, Université François-Rabelais, Tours, France
| | - Jayne M Christen
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - David F Clarke
- CSIRO Land and Water, Clunies Ross St, Acton, ACT, 2601, Australia
| | - Neal T Dittmer
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | | | - Spyridoula Garavelou
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Karl H J Gordon
- CSIRO Health and Biosecurity, Clunies Ross St, Acton, ACT, 2601, Australia
| | - Ramesh T Gunaratna
- Department of Entomology and Plant Pathology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Yi Han
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Frank Hauser
- Center for Functional and Comparative Insect Genomics, Department of Biology, University of Copenhagen, Universitetsparken 15, DK-21oo, Copenhagen, Denmark
| | - Yan He
- Department of Entomology and Plant Pathology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Hanna Heidel-Fischer
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - Ariana Hirsh
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| | - Yingxia Hu
- Department of Entomology and Plant Pathology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Hongbo Jiang
- Key Laboratory of Entomology and Pest Control Engineering, College of Plant Protection, Southwest University, Chongqing, 400715, China
| | - Divya Kalra
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Christian Klinner
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knoell-Strasse, 8, D-07745, Jena, Germany
| | - Christopher König
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knoell-Strasse, 8, D-07745, Jena, Germany
| | - Christie Kovar
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Ashley R Kroll
- Department of Biology, Reed College, Portland, OR, 97202, USA
| | - Suyog S Kuwar
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - Sandy L Lee
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Rüdiger Lehman
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Bioresources Project Group, Winchesterstrasse 2, 35394, Gießen, Germany
| | - Kai Li
- College of Chemistry, Chemical Engineering, and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Zhaofei Li
- College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Hanquan Liang
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Shanna Lovelace
- Department of Biological Sciences, University of Southern Maine, Portland, ME, 04104, USA
| | - Zhiqiang Lu
- College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jennifer H Mansfield
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| | - Kyle J McCulloch
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, 92697, USA
| | - Tittu Mathew
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Brian Morton
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - David Neunemann
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - Fiona Ongeri
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Yannick Pauchet
- Department of Entomology, Max Planck Institute for Chemical Ecology, Hans-Knoell-Strasse 8, 07745, Jena, Germany
| | - Ling-Ling Pu
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Ioannis Pyrousis
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Xiang-Jun Rao
- School of Plant Protection, Anhui Agricultural University, Hefei, Anhui, China
| | - Amanda Redding
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Charles Roesel
- Department of Marine and Environmental Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Alejandro Sanchez-Gracia
- Departament de Genètica and Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Barcelona, Spain
| | - Sarah Schaack
- Department of Biology, Reed College, Portland, OR, 97202, USA
| | - Aditi Shukla
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY, 10027, USA
| | - Guillaume Tetreau
- Department of Entomology, Cornell University, New York State Agricultural Experiment Station, Geneva, NY, 14456, USA
| | - Yang Wang
- Department of Entomology and Plant Pathology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Guang-Hua Xiong
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Walther Traut
- Institut fuer Biologie, Universitaet Luebeck, D-23538, Luebeck, Germany
| | - Tom K Walsh
- CSIRO Land and Water, Clunies Ross St, Acton, ACT, 2601, Australia
| | - Kim C Worley
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Di Wu
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - Wenbi Wu
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Yuan-Qing Wu
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Xiufeng Zhang
- Department of Entomology and Plant Pathology, Oklahoma State University, Stillwater, OK, 74078, USA
| | - Zhen Zou
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hannah Zucker
- Neuroscience Program, Hamilton College, Clinton, NY, 13323, USA
| | - Adriana D Briscoe
- Department of Ecology and Evolutionary Biology, University of California, Irvine, CA, 92697, USA
| | | | - Rollie J Clem
- Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - René Feyereisen
- Department of Crop Protection, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Cornelis J P Grimmelikhuijzen
- Center for Functional and Comparative Insect Genomics, Department of Biology, University of Copenhagen, Universitetsparken 15, DK-21oo, Copenhagen, Denmark
| | - Stavros J Hamodrakas
- Department of Cell Biology and Biophysics, Faculty of Biology, University of Athens, Athens, Greece
| | - Bill S Hansson
- Max Planck Institute for Chemical Ecology, Department of Evolutionary Neuroethology, Hans-Knoell-Strasse, 8, D-07745, Jena, Germany
| | - Elisabeth Huguet
- Institut de Recherche sur la Biologie de l'Insecte, UMR CNRS 7261, UFR Sciences et Techniques, Université François-Rabelais, Tours, France
| | - Lars S Jermiin
- CSIRO Land and Water, Clunies Ross St, Acton, ACT, 2601, Australia
| | - Que Lan
- Department of Entomology, University of Wisconsin, Madison, USA
| | - Herman K Lehman
- Biology Department and Neuroscience Program, Hamilton College, Clinton, NY, 13323, USA
| | - Marce Lorenzen
- Dept. Entomology, North Carolina State Univ., Raleigh, NC, 27695, USA
| | - Hans Merzendorfer
- University of Siegen, School of Natural Sciences and Engineering, Institute of Biology - Molecular Biology, Adolf-Reichwein-Strasse. 2, AR-C3010, 57076 Siegen, Germany
| | - Ioannis Michalopoulos
- Centre of Systems Biology, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - David B Morton
- Department of Integrative Biosciences, School of Dentistry, BRB421, L595, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Subbaratnam Muthukrishnan
- Department of Biochemistry and Molecular Biophysics, Kansas State University, Manhattan, KS, 66506, USA
| | - John G Oakeshott
- CSIRO Land and Water, Clunies Ross St, Acton, ACT, 2601, Australia
| | - Will Palmer
- Department of Genetics, University of Cambridge, Downing St, Cambridge, CB2 3EH, UK
| | - Yoonseong Park
- Department of Entomology, Kansas State University, Manhattan, KS, 66506, USA
| | | | - Julio Rozas
- Departament de Genètica and Institut de Recerca de la Biodiversitat (IRBio), Universitat de Barcelona, Barcelona, Spain
| | | | - Wendy Smith
- Department of Biology, Northeastern University, Boston, MA, 02115, USA
| | - Agnes Southgate
- Department of Biology, College of Charleston, Charleston, SC, 29424, USA
| | - Andreas Vilcinskas
- Institute for Insect Biotechnology, Justus-Liebig-University, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Richard Vogt
- Department of Biological Sciences, University of South Carolina, Columbia, SC, 29205, USA
| | - Ping Wang
- Department of Entomology, Cornell University, New York State Agricultural Experiment Station, Geneva, NY, 14456, USA
| | - John Werren
- Department of Biology, University of Rochester, Rochester, NY, 14627, USA
| | - Xiao-Qiang Yu
- University of Missouri-Kansas City, 5007 Rockhill Road, Kansas City, MO, 64110, USA
| | - Jing-Jiang Zhou
- Department of Biological Chemistry and Crop Protection, Rothamsted Research, Harpenden, Herts, AL5 2JQ, UK
| | - Susan J Brown
- KSU Bioinformatics Center, Division of Biology, Kansas State University, Manhattan, KS, 66506, USA
| | - Steven E Scherer
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Stephen Richards
- Human Genome Sequencing Center, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Gary W Blissard
- Boyce Thompson Institute at Cornell University, Tower Road, Ithaca, NY, 14853, USA
| |
Collapse
|
46
|
Sieber MH, Thomsen MB, Spradling AC. Electron Transport Chain Remodeling by GSK3 during Oogenesis Connects Nutrient State to Reproduction. Cell 2016; 164:420-32. [PMID: 26824655 PMCID: PMC6894174 DOI: 10.1016/j.cell.2015.12.020] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 10/30/2015] [Accepted: 11/26/2015] [Indexed: 11/28/2022]
Abstract
Reproduction is heavily influenced by nutrition and metabolic state. Many common reproductive disorders in humans are associated with diabetes and metabolic syndrome. We characterized the metabolic mechanisms that support oogenesis and found that mitochondria in mature Drosophila oocytes enter a low-activity state of respiratory quiescence by remodeling the electron transport chain (ETC). This shift in mitochondrial function leads to extensive glycogen accumulation late in oogenesis and is required for the developmental competence of the oocyte. Decreased insulin signaling initiates ETC remodeling and mitochondrial respiratory quiescence through glycogen synthase kinase 3 (GSK3). Intriguingly, we observed similar ETC remodeling and glycogen uptake in maturing Xenopus oocytes, suggesting that these processes are evolutionarily conserved aspects of oocyte development. Our studies reveal an important link between metabolism and oocyte maturation.
Collapse
Affiliation(s)
- Matthew H Sieber
- Department of Embryology, Carnegie Institution of Washington, 3520 San Martin Drive, Baltimore, MD 21218, USA
| | - Michael B Thomsen
- Department of Biology, Johns Hopkins University, 3400 North Charles Street, Baltimore, MD 21218, USA
| | - Allan C Spradling
- Department of Embryology, Carnegie Institution of Washington, 3520 San Martin Drive, Baltimore, MD 21218, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
47
|
Tschapalda K, Zhang YQ, Liu L, Golovnina K, Schlemper T, Eichmann TO, Lal-Nag M, Sreenivasan U, McLenithan J, Ziegler S, Sztalryd C, Lass A, Auld D, Oliver B, Waldmann H, Li Z, Shen M, Boxer MB, Beller M. A Class of Diacylglycerol Acyltransferase 1 Inhibitors Identified by a Combination of Phenotypic High-throughput Screening, Genomics, and Genetics. EBioMedicine 2016; 8:49-59. [PMID: 27428418 PMCID: PMC4919474 DOI: 10.1016/j.ebiom.2016.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/02/2016] [Accepted: 04/12/2016] [Indexed: 12/13/2022] Open
Abstract
Excess lipid storage is an epidemic problem in human populations. Thus, the identification of small molecules to treat or prevent lipid storage-related metabolic complications is of great interest. Here we screened > 320.000 compounds for their ability to prevent a cellular lipid accumulation phenotype. We used fly cells because the multifarious tools available for this organism should facilitate unraveling the mechanism-of-action of active small molecules. Of the several hundred lipid storage inhibitors identified in the primary screen we concentrated on three structurally diverse and potent compound classes active in cells of multiple species (including human) and negligible cytotoxicity. Together with Drosophila in vivo epistasis experiments, RNA-Seq expression profiles suggested that the target of one of the small molecules was diacylglycerol acyltransferase 1 (DGAT1), a key enzyme in the production of triacylglycerols and prominent human drug target. We confirmed this prediction by biochemical and enzymatic activity tests. We identified > 600 potent small molecule inhibitors of cellular lipid storage deposition. RNA-Seq expression profiling discriminated the activity of three lead scaffolds and guided subsequent functional studies. We discovered a class of DGAT1 inhibitors, which is active in fly and mammalian cell lines as well as whole flies.
Obesity and other lipid storage associated diseases are a growing health threat of human populations. In an undirected phenotypic screen, we identified pharmacologically active small molecules that reduce or enhance lipid storage. Our work focuses on three lead structures that prevent lipid storage in diverse cellular systems including cells from a diabetes patient. In order to elucidate the compound mechanisms-of-action and cellular targets, we used a combination of RNA-Seq transcriptional profiling and diverse functional assays. Our results strongly suggest that one of our lead structures represents a class of inhibitors targeting the key lipogenic enzyme diacylglycerol acyltransferase 1.
Collapse
Affiliation(s)
- Kirsten Tschapalda
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Germany; Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Germany; Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany; Department of Molecular Developmental Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Ya-Qin Zhang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Li Liu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Kseniya Golovnina
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, USA
| | - Thomas Schlemper
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Germany; Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Germany
| | | | - Madhu Lal-Nag
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Urmila Sreenivasan
- Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA
| | - John McLenithan
- Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA
| | - Slava Ziegler
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Carole Sztalryd
- Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA; Baltimore VA Medical Center, VA Research Service, Geriatric Research, Education and Clinical Center (GRECC) and VA Maryland Health Care System, Department of Medicine, Division of Endocrinology University of Maryland School of Medicine, USA
| | - Achim Lass
- Institute of Molecular Biosciences, University of Graz, Austria
| | - Douglas Auld
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Brian Oliver
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, USA
| | - Herbert Waldmann
- Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Zhuyin Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Min Shen
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Matthew B Boxer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, USA
| | - Mathias Beller
- Systems Biology of Lipid Metabolism, Heinrich Heine University Düsseldorf, Germany; Institute for Mathematical Modeling of Biological Systems, Heinrich Heine University Düsseldorf, Germany.
| |
Collapse
|
48
|
Soulages JL, Wu Z, Firdaus SJ, Mahalingam R, Arrese EL. Monoacylglycerol and diacylglycerol acyltransferases and the synthesis of neutral glycerides in Manduca sexta. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 62:194-210. [PMID: 25263765 PMCID: PMC4377122 DOI: 10.1016/j.ibmb.2014.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Revised: 09/09/2014] [Accepted: 09/15/2014] [Indexed: 05/06/2023]
Abstract
The insect fat body and the adipose tissue of vertebrates store fatty acids (FA) as triacylglycerols (TG). However, the fat body of most insects has the unique ability to rapidly produce and secrete large amounts of diacylglycerol (DG). Monoacylglycerol acyltransferase (MGAT), which catalyzes the synthesis of DG from MG, and a diacylglycerol acyltransferase (DGAT), which catalyzes the synthesis of TG from DG, are key enzymes in the metabolism of neutral glycerides. However, very little is known about these acyltransferases in insects. In the present study we have cloned two predicted MGATs and a DGAT from Manduca sexta and compared their sequences with predicted MGAT and DGAT homologs from a number of insect species. The comparison suggested that insects may only have a single DGAT gene, DGAT1. The apparent absence of a DGAT2 gene in insects would represent a major difference with vertebrates, which contain DGAT1 and DGAT2 genes. Insects seem to have a single MGAT gene which is similar to the MGAT2 of vertebrates. A number of conserved phosphorylation sites of potential physiological significance were identified among insect proteins and among insect and vertebrate proteins. DGAT1 and MGAT are expressed in fat body, midgut and ovaries. The relative rates of utilization of FAs for the synthesis of DG and TG correlated with the relative expression levels of MGAT and DGAT suggesting that regulation of the expression levels of these acyltransferases could be determining whether the fat body secretes DG or stores fatty acids as TG. The expression patterns of the acyltransferases suggest a role of the monoacylglycerol pathway in the production and mobilization of DG in M. sexta fat body.
Collapse
Affiliation(s)
- Jose L Soulages
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA.
| | - Zengying Wu
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Sarah J Firdaus
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Ramamurthy Mahalingam
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA
| | - Estela L Arrese
- Department of Biochemistry and Molecular Biology, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
49
|
Welte MA. As the fat flies: The dynamic lipid droplets of Drosophila embryos. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:1156-85. [PMID: 25882628 DOI: 10.1016/j.bbalip.2015.04.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 03/31/2015] [Accepted: 04/06/2015] [Indexed: 01/09/2023]
Abstract
Research into lipid droplets is rapidly expanding, and new cellular and organismal roles for these lipid-storage organelles are continually being discovered. The early Drosophila embryo is particularly well suited for addressing certain questions in lipid-droplet biology and combines technical advantages with unique biological phenomena. This review summarizes key features of this experimental system and the techniques available to study it, in order to make it accessible to researchers outside this field. It then describes the two topics most heavily studied in this system, lipid-droplet motility and protein sequestration on droplets, discusses what is known about the molecular players involved, points to open questions, and compares the results from Drosophila embryo studies to what it is known about lipid droplets in other systems.
Collapse
Affiliation(s)
- Michael A Welte
- Department of Biology University of Rochester, RC Box 270211, 317 Hutchison Hall, Rochester, NY 14627, USA.
| |
Collapse
|
50
|
Steroid Signaling Establishes a Female Metabolic State and Regulates SREBP to Control Oocyte Lipid Accumulation. Curr Biol 2015; 25:993-1004. [PMID: 25802149 PMCID: PMC6894397 DOI: 10.1016/j.cub.2015.02.019] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/12/2014] [Accepted: 02/03/2015] [Indexed: 11/21/2022]
Abstract
Disruptions in energy homeostasis severely affect reproduction in many organisms and are linked to several reproductive disorders in humans. As a result, understanding the mechanisms that control nutrient accumulation in the oocyte will provide valuable insights into the links between metabolic disease and reproductive dysfunction. We show that the steroid hormone ecdysone functions in Drosophila to control lipid metabolism and support oocyte production. First, local EcR-mediated signaling induces a stage-specific accumulation of lipids in stage-10 oocytes. EcR induces lipid accumulation by promoting the activation of the lipogenic transcription factor SREBP and by controlling the expression of the low-density lipoprotein (LDL) receptor homolog, LpR2. Second, global signaling via the ecdysone receptor, EcR, establishes a female metabolic state and promotes whole-body triglyceride and glycogen storage at high levels. EcR acts in the CNS to mediate these effects, in part by promoting higher levels of feeding in females. Thus, ecdysone functions at two levels to support reproduction: first by inducing lipid accumulation in the late stages of oocyte development and second by providing a signal that coordinates lipid metabolism in the germline with whole-animal lipid homeostasis. Ecdysone regulation allows females to assess the demands of oogenesis and alter their behavior and metabolic state to support the biosynthetic requirements of oocyte production.
Collapse
|