1
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
2
|
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
|
3
|
Diaz AV, Tekin I, Reis T. Drosophila as a Genetic Model System to Study Organismal Energy Metabolism. Biomolecules 2025; 15:652. [PMID: 40427545 PMCID: PMC12108566 DOI: 10.3390/biom15050652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolism is the essential process by which an organism converts nutrients into energy to fuel growth, development, and repair. Metabolism at the level of a multicellular, multi-organ animal is inherently more complex than metabolism at the single-cell level. Indeed, each organ also must maintain its own homeostasis to function. At all three scales, homeostasis is a defining feature: as energy sources and energetic demands wax and wane, the system must be robust. While disruption of organismal energy homeostasis can be manifested in different ways in humans, obesity (defined as excess body fat) is an increasingly common outcome of metabolic imbalance. Here we will discuss the genetic basis of metabolic dysfunction that underlies obesity. We focus on what we are learning from Drosophila melanogaster as a model organism to explore and dissect genetic causes of metabolic dysfunction in the context of a whole organism.
Collapse
Affiliation(s)
| | | | - Tânia Reis
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
4
|
Wang Q, Liu JL, Liu J. CTPS cytoophidia in Drosophila: distribution, regulation, and physiological roles. Exp Cell Res 2025; 447:114536. [PMID: 40122502 DOI: 10.1016/j.yexcr.2025.114536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 03/25/2025]
Abstract
Intracellular compartmentalization plays a critical role in maintaining cellular homeostasis and regulating metabolic processes. A growing body of evidence suggests that various metabolic enzymes, including CTP synthase (CTPS), can dynamically assemble into membraneless filamentous structures. The formation of these membraneless organelles is precisely regulated by the cellular metabolic state. CTPS, a rate-limiting enzyme in the de novo biosynthesis of CTP, has been shown to assemble into filamentous structures known as cytoophidium. First identified in 2010 by three independent research groups, cytoophidia are evolutionarily conserved across diverse organisms, including bacteria, archaea, yeast, mammals, and plants, suggesting a fundamental biological function. Given the well-established advantages of Drosophila melanogaster as a genetic model, this organism provides a powerful system for investigating the physiological roles of cytoophidia. This review synthesizes current findings on CTPS cytoophidia in Drosophila, with a particular focus on their spatiotemporal distribution in tissues and their regulatory roles in three key biological processes: intestinal homeostasis, lipid metabolism, and reproductive physiology. Furthermore, we discuss the challenges and future directions in cytoophidia research, offering insights into their broader implications in cellular metabolism and physiology.
Collapse
Affiliation(s)
- Qingyi Wang
- College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Jingnan Liu
- College of Life Sciences, Shanghai Normal University, Shanghai, 200234, China.
| |
Collapse
|
5
|
Johnson MG, Barrett M. Review: Exploring correctness, usefulness, and feasibility of potential physiological operational welfare indicators for farmed insects to establish research priorities. Animal 2025:101501. [PMID: 40288947 DOI: 10.1016/j.animal.2025.101501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
While insects are already the largest group of terrestrial food and feed livestock animals in terms of absolute number of individuals, the insect farming industry is expected to continue growing rapidly in order to meet the nutritional demands of the human population during the 21st century. Accordingly, consumers, producers, legislators, and industry-adjacent researchers have expressed interest in further research and assessment of farmed insect welfare. Operational indicators of animal welfare are those that can be used to putatively assess the welfare of animals in the absence of true indicators of affective state (e.g., valenced/emotional state) and are commonly used for farmed vertebrate livestock species; however, significant behavioral and physiological differences between vertebrates and insects means these indicators must be examined for their correctness, usefulness, and feasibility prior to use with insect livestock. The most valuable operational welfare indicators would (1) correctly correspond to the insect's putative welfare state; (2) provide useful information about what is affecting the insect's welfare; and (3) be feasible for deployment at a large scale on farms. As there are many possible indicators that could be further researched in insects, evaluating the likely correctness, feasibility, and usefulness of these indicators in insects will allow researchers to prioritize which indicators to investigate first for use on farms. Thus, in this review, we explore whether physiological or somatic indicators of farmed vertebrate welfare, including whole-body, immune, neurobiological, and respiratory/cardiac indicators, may be correct, feasible, and useful for assessing farmed insect welfare. We review insect physiological systems, as well as any existing, welfare-relevant data from farmed or closely related insects. We end by proposing a priority list for physiological, operational welfare indicators that are most likely to correctly, usefully, and feasibly assess farmed insect welfare, which may guide indicator validation research priorities for insect welfare scientists.
Collapse
Affiliation(s)
- M G Johnson
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58108, USA
| | - M Barrett
- Department of Biology, Indiana University Indianapolis, Indianapolis, IN 46202 USA.
| |
Collapse
|
6
|
Meng TK, Han RL, Ma P, Chen SX, Qi BH, Wang ZX, Li XY, Deng HS. Microemulsion-based drug delivery system identifies pepper alkaloids as anti-obesity compounds. Acta Pharmacol Sin 2025:10.1038/s41401-025-01521-x. [PMID: 40113987 DOI: 10.1038/s41401-025-01521-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/23/2025] [Indexed: 03/22/2025]
Abstract
Obesity is a significant contributor to various metabolic diseases such as heart disease and diabetes. Due to the adverse effects of synthetic anti-obesity drugs, natural products from functional food plants, which mimic the effects of synthetic chemicals, present promising alternatives. However, many natural plant-derived compounds are poorly soluble in water, resulting in low bioavailability within the gastrointestinal tract, a key limitation for the effectiveness of many hydrophobic substances. In this study we developed a microemulsion-based drug delivery system in Drosophila, which effectively enhanced the solubility of hydrophobic compounds without noticeable effects on food intake or survival in fruit flies. This system consisted of cremophor EL, ethanol and ethyl oleate (7:6:1), which enabled the establishment of an emulsion-based liquid high-fat diet (LHFD) model, followed by a pilot screening of 161 standard substances from traditional Chinese medicine. We found that piperine (PIP), an alkaloid derived from black pepper, significantly decreased triacylglycerol (TAG) levels in both the intestine and in whole flies. We demonstrated that piperine (1 mg/ml) significantly elevated cytosolic Ca2+ levels in enterocytes by activating Transient receptor potential (TRP) channels. TRPV1 agonists such as capsaicin and evodiamine (another alkaloid identified during the screening) also exhibited anti-obesity effects. Increased Ca2+ levels resulted in the suppression of dietary lipase Magro expression through the activation of the transcription factor cAMP response element binding protein (CREB). Furthermore, hydrophobic compounds in the microemulsion were successfully delivered to distal tissues including liver and brain blood vessels in mice, and PIP in the microemulsion was sufficient to reduce body weight in mice. In conclusion, we have developed a microemulsion-based U-GLAD platform for drug delivery, and piperine is identified as a weight-controlling compound, providing a novel approach to the treatment of obesity and its associated symptoms.
Collapse
Affiliation(s)
- Tian-Kai Meng
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Ruo-Lei Han
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Peng Ma
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shu-Xin Chen
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bo-Han Qi
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zi-Xuan Wang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Xiao-Yu Li
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Han-Song Deng
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
7
|
Zhao Y, Duan J, van de Leemput J, Han Z. Cardiac neurons expressing a glucagon-like receptor mediate cardiac arrhythmia induced by high-fat diet in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.13.571403. [PMID: 40161619 PMCID: PMC11952361 DOI: 10.1101/2023.12.13.571403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Cardiac arrhythmia leads to increased risks for stroke, heart failure, and cardiac arrest. Arrhythmic pathology is often rooted in the cardiac conduction system, but the mechanism is complex and not fully understood. For example, how metabolic diseases, like obesity and diabetes, increase the risk for cardiac arrhythmia. Glucagon regulates glucose production, mobilizes lipids from the fat body, and affects cardiac rate and rhythm, attributes of a likely key player. Drosophila is an established model to study metabolic diseases and cardiac arrhythmias. Since glucagon signaling is highly conserved, we used high-fat diet (HFD)-fed flies to study its effect on heart function. HFD led to increased heartbeat and an irregular rhythm. The HFD-fed flies showed increased levels of adipokinetic hormone (Akh), the functional equivalent to human glucagon. Both genetic reduction of Akh and eliminating the Akh producing cells (APC) rescued HFD-induced arrhythmia, whereas heart rhythm was normal in Akh receptor mutants (AkhRnull ). Furthermore, we discovered a pair of cardiac neurons that express high levels of Akh receptor. These are located near the posterior heart, make synaptic connections at the heart muscle, and regulate heart rhythm. Altogether, this Akh signaling pathway provides new understanding of the regulatory mechanisms between metabolic disease and cardiac arrhythmia.
Collapse
Affiliation(s)
- Yunpo Zhao
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jianli Duan
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
8
|
Akagi K, Jin YJ, Koizumi K, Oku M, Ito K, Shen X, Imura JI, Aihara K, Saito S. Integration of Dynamical Network Biomarkers, Control Theory and Drosophila Model Identifies Vasa/DDX4 as the Potential Therapeutic Targets for Metabolic Syndrome. Cells 2025; 14:415. [PMID: 40136664 PMCID: PMC11941168 DOI: 10.3390/cells14060415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic syndrome (MetS) is a subclinical disease, resulting in increased risk of type 2 diabetes (T2D), cardiovascular diseases, cancer, and mortality. Dynamical network biomarkers (DNB) theory has been developed to provide early-warning signals of the disease state during a preclinical stage. To improve the efficiency of DNB analysis for the target genes discovery, the DNB intervention analysis based on the control theory has been proposed. However, its biological validation in a specific disease such as MetS remains unexplored. Herein, we identified eight candidate genes from adipose tissue of MetS model mice at the preclinical stage by the DNB intervention analysis. Using Drosophila, we conducted RNAi-mediated knockdown screening of these candidate genes and identified vasa (also known as DDX4), encoding a DEAD-box RNA helicase, as a fat metabolism-associated gene. Fat body-specific knockdown of vasa abrogated high-fat diet (HFD)-induced enhancement of starvation resistance through up-regulation of triglyceride lipase. We also confirmed that DDX4 expressing adipocytes are increased in HFD-fed mice and high BMI patients using the public datasets. These results prove the potential of the DNB intervention analysis to search the therapeutic targets for diseases at the preclinical stage.
Collapse
Affiliation(s)
- Kazutaka Akagi
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan;
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Ying-Jie Jin
- Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-0194, Japan;
| | - Keiichi Koizumi
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan;
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Makito Oku
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Kaisei Ito
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan;
| | - Xun Shen
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan;
| | - Jun-ichi Imura
- Department of Systems and Control Engineering, School of Engineering, Institute of Science Tokyo, Tokyo 152-8552, Japan;
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo 113-0033, Japan;
| | - Shigeru Saito
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| |
Collapse
|
9
|
Chikamatsu S, Sakakibara Y, Takei K, Nishijima R, Iijima KM, Sekiya M. Supplementation of essential amino acids suppresses age-associated sleep loss and sleep fragmentation but not loss of rhythm strength under yeast-restricted malnutrition in Drosophila. J Biochem 2025; 177:225-237. [PMID: 39696747 PMCID: PMC11879319 DOI: 10.1093/jb/mvae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/12/2024] [Accepted: 12/16/2024] [Indexed: 12/20/2024] Open
Abstract
Sleep quality and quantity decrease with age, and sleep disturbance increases the risk of many age-associated diseases. There is a significant relationship between nutritional status and sleep outcomes, with malnutrition inducing poor sleep quality in older adults. However, it remains elusive whether, and if so how, nutritional supplementation prevents age-associated sleep problems. Here, we utilized Drosophila to investigate the effects of a malnutrition diet with restricted yeast, a primary protein source, and supplementation of 10 essential amino acids (EAAs) on sleep profiles during ageing. Compared with the standard diet containing 2.7% yeast, the malnutrition diet containing 0.27% yeast significantly decreased target of rapamycin (TOR) signalling and shortened the lifespan of male Canton-S flies. By contrast, age-associated sleep loss, sleep fragmentation and loss of rhythm strength were similarly observed under both diets. Supplementation of the malnutrition diet with EAAs in restricted yeast significantly ameliorated age-associated sleep loss and sleep fragmentation without altering loss of rhythm strength. It also rescued decreased TOR signalling activity but not the shortened lifespan, suggesting that the effects of EAAs on sleep integrity are independent of TOR activity and lifespan regulation. These results may help to develop dietary interventions that improve age-related sleep problems in humans.
Collapse
Affiliation(s)
- Sachie Chikamatsu
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603 Japan
| | - Yasufumi Sakakibara
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
| | - Kimi Takei
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
| | - Risa Nishijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
| | - Koichi M Iijima
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603 Japan
| | - Michiko Sekiya
- Department of Neurogenetics, Center for Development of Advanced Medicine for Dementia, National Center for Geriatrics and Gerontology, 7-430 Morioka-cho, Obu, Aichi 474-8511, Japan
- Department of Experimental Gerontology, Graduate School of Pharmaceutical Sciences, Nagoya City University, 3-1 Tanabe-dori, Mizuho-ku, Nagoya, Aichi 467-8603 Japan
| |
Collapse
|
10
|
Mercola BM, Villalobos TV, Wood JE, Basu A, Johnson AE. Increased expression of the small lysosomal gene SVIP in the Drosophila gut suppresses pathophysiological features associated with a high-fat diet. Biol Open 2025; 14:BIO061601. [PMID: 39882732 PMCID: PMC11810118 DOI: 10.1242/bio.061601] [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: 06/19/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025] Open
Abstract
Lysosomes are digestive organelles that are crucial for nutrient sensing and metabolism. Lysosome impairment is linked to a broad spectrum of metabolic disorders, underscoring their importance to human health. Thus, lysosomes are an attractive target for metabolic disease therapies. In previous work, we discovered a novel class of tubular lysosomes that are morphologically and functionally distinct from traditionally described vesicular lysosomes. Tubular lysosomes are present in multiple tissues, are broadly conserved from invertebrates to mammals, are more proficient at degrading autophagic cargo than vesicular lysosomes, and delay signs of tissue aging when induced ectopically. Thus, triggering tubular lysosome formation presents one mechanism to increase lysosome activity and, notably, overproduction of the small lysosomal protein, SVIP, is a robust genetic strategy for triggering lysosomal tubulation on demand. In this study, we examine whether SVIP overexpression in the fly gut can suppress pathophysiological phenotypes associated with an obesogenic high-fat diet. Indeed, our results indicate that increasing SVIP expression in the fly gut reduces lipid accumulation, suppresses body mass increase, and improves survival in flies fed a high-fat diet. Collectively, these data hint that increasing lysosomal activity through induction of tubular lysosomal networks, could be one strategy to combat obesity-related pathologies.
Collapse
Affiliation(s)
- Brennan M. Mercola
- Louisiana State University, Department of Biological Sciences, Baton Rouge, LA 70803, USA
| | - Tatiana V. Villalobos
- Louisiana State University, Department of Biological Sciences, Baton Rouge, LA 70803, USA
| | - Jocelyn E. Wood
- Louisiana State University, Department of Biological Sciences, Baton Rouge, LA 70803, USA
| | - Ankita Basu
- Louisiana State University, Department of Biological Sciences, Baton Rouge, LA 70803, USA
| | - Alyssa E. Johnson
- Louisiana State University, Department of Biological Sciences, Baton Rouge, LA 70803, USA
| |
Collapse
|
11
|
Wellings JJ, Thorpe JM, Yendole K, Matsubayashi Y, Hartley PS. Effect of short and long-term cadmium exposure on behaviour and cardiac function in Drosophila. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125481. [PMID: 39644948 DOI: 10.1016/j.envpol.2024.125481] [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: 09/26/2024] [Revised: 11/17/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Cadmium is a non-essential heavy metal and environmental pollutant that causes a range of pathologies across different species. In humans, cadmium exposure has recently been directly linked to heart disease. Understanding how long-term cadmium exposure affects cardiac physiology is therefore important. In this work we employed a tractable Drosophila melanogaster model to study the effects of cadmium exposure on behaviour, lifespan and cardiac physiology. Dietary experiments established that cadmium at 10 μM and 100 μM was tolerated for several weeks, whereas doses in the mM range caused lethality within days. It was estimated that 10 μM dietary exposure represented an approximately 60-fold excess of the maximum exposure recommended for humans. Although 10 μM cadmium had no impact on lifespan compared to the control diet, it did cause significant daytime hyperactivity. Direct exposure of the heart to cadmium caused reversible cardiac arrest and disrupted calcium signalling. Compared to controls, 10 μM dietary cadmium had no impact on the rate of cardiac ageing over a six-week period. The higher dose of 100 μM shortened the flies' lifespan but it slowed the rate of cardiac ageing. The findings indicate that Drosophila can be used to model the direct effects of cadmium on cardiomyocyte function and also demonstrate the existence of cardioprotective pathways triggered by dietary cadmium exposure. The data also indicate that cadmium at doses that do not affect lifespan or heart function, do cause daytime hyperactivity. Identifying the cardioprotective mechanisms of cadmium and understanding the hyperactivity phenotype in Drosophila may yield important findings of applied relevance to insects in general, as well as humans exposed to cadmium.
Collapse
Affiliation(s)
- Jessica J Wellings
- Department of Life and Environmental Science, Bournemouth University, Poole, Dorset, BH12 5BB, UK
| | - Jamie M Thorpe
- Department of Life and Environmental Science, Bournemouth University, Poole, Dorset, BH12 5BB, UK
| | - Karen Yendole
- Department of Life and Environmental Science, Bournemouth University, Poole, Dorset, BH12 5BB, UK
| | - Yutaka Matsubayashi
- Department of Life and Environmental Science, Bournemouth University, Poole, Dorset, BH12 5BB, UK
| | - Paul S Hartley
- Department of Life and Environmental Science, Bournemouth University, Poole, Dorset, BH12 5BB, UK.
| |
Collapse
|
12
|
Obafemi OT, Ayeleso AO, Adewale OB, Unuofin J, Ekundayo BE, Ntwasa M, Lebelo SL. Animal models in biomedical research: Relevance of Drosophila melanogaster. Heliyon 2025; 11:e41605. [PMID: 39850441 PMCID: PMC11754520 DOI: 10.1016/j.heliyon.2024.e41605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 12/23/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Animal models have become veritable tools in gaining insight into the pathogenesis and progression of several human diseases. These models could range in complexity from Caenorhabditis elegans to non-human primates. With the aid of these animal models, a lot of new knowledge has been gained about several diseases which otherwise would not have been possible. Most times, the utilization of these animal models is predicated on the level of homology they share with humans, which suggests that outcomes of studies using them could be extrapolated to humans. However, this has not always been the case. Drosophila melanogaster is becoming increasingly relevant as preferred model for understanding the biochemical basis of several human diseases. Apart from its relatively short lifespan, high fecundity and ease of rearing, the simplicity of its genome and lower redundancy of its genes when compared with vertebrate models, as well as availability of genetic tool kit for easy manipulation of its genome, have all contributed to its emergence as a valid animal model of human diseases. This review aimed at highlighting the contributions of selected animal models in biomedical research with a focus on the relevance of Drosophila melanogaster in understanding the biochemical basis of some diseases that have continued to plague mankind.
Collapse
Affiliation(s)
- Olabisi Tajudeen Obafemi
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | - Ademola Olabode Ayeleso
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
- Biochemistry Programme, College of Agriculture, Engineering and Science, Bowen University, PMB 284, Iwo, Osun State, Nigeria
| | | | - Jeremiah Unuofin
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | | | - Monde Ntwasa
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| | - Sogolo Lucky Lebelo
- Department of Life and Consumer Sciences, School of Agriculture and Life Sciences, University of South Africa, 1710, Johannesburg, South Africa
| |
Collapse
|
13
|
Sahu S, Dash K, Mishra M. Common salt (NaCl) causes developmental, behavioral, and physiological defects in Drosophila melanogaster. Nutr Neurosci 2025:1-19. [PMID: 39760749 DOI: 10.1080/1028415x.2024.2441677] [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: 01/07/2025]
Abstract
PURPOSE The incidence of obesity has surged to pandemic levels in recent decades. Approximately 1.89 million obesity are linked to excessive salt consumption. This study aims to check the toxicity of salt at different concentrations using an invertebrate model organism Drosophila melanogaster. METHODS Drosophila food was mixed with different salt concentrations (50, 200, 400, 800 µM). The toxicity of salt in third instar larvae was checked via different experiments such as trypan blue assay, crawling assay, and other histological staining was done to check the deposition of lipid droplets and amount of reactive oxygen species. Food intake analysis was performed to check the feeding rate, and body weight was also calculated to check the obesity index. Several behavioral assays are also performed in adult flies. RESULTS Most significant abnormalities were seen at 50 and 200 µM concentrations. Feeding rate increased up to 60%, body weight was increased up to 12% in larvae, and 27% in adult at 200 µM concentration. Approximately 60% larvae and 58% adult flies had defective response to extreme heat. 28% larvae and 38% adult flies were not responding to cold temperature. 55% flies had a defective phototaxis behavior and 40% of them showed positive geotaxis at those range. Salt stress leads to the buildup of free radicals, resulting in DNA damage in both the gut and hemolymph. FINDINGS Most toxic consequences are observed at the lower concentration range as the feeding rate was higher. Flies show aversive response to feed on the higher concentration of salt.
Collapse
Affiliation(s)
- Swetapadma Sahu
- Neural Developmental Biology Lab, Department of Life Science, NIT Rourkela, Rourkela, Odisha, India
| | - Kalpanarani Dash
- Neural Developmental Biology Lab, Department of Life Science, NIT Rourkela, Rourkela, Odisha, India
| | - Monalisa Mishra
- Neural Developmental Biology Lab, Department of Life Science, NIT Rourkela, Rourkela, Odisha, India
| |
Collapse
|
14
|
Zhu L, Xie Y, Liu C, Cheng J, Shen Z, Liu X, Cai L, Ning X, Zhang S, Li Z, Huang Q, Liu X. Baculoviruses manipulate host lipid metabolism via adipokinetic hormone signaling to induce climbing behavior. PLoS Pathog 2025; 21:e1012932. [PMID: 39888969 PMCID: PMC11819524 DOI: 10.1371/journal.ppat.1012932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 02/12/2025] [Accepted: 01/23/2025] [Indexed: 02/02/2025] Open
Abstract
Baculoviruses can induce climbing behavior in caterpillar hosts, which provides an excellent model for studying parasite manipulation of host behavior. Herein, we found that Helicoverpa armigera single nucleopolyhedrovirus (HearNPV) promoted lipid metabolism of infected H. armigera larvae, and changes in lipid metabolism can affect climbing behavior. Therefore, understanding the molecular mechanisms between lipid metabolism and climbing behavior is particularly important. In this study, we found adipokinetic hormone 1 (HaAKH1), adipokinetic hormone 2 (HaAKH2) and their receptor HaAKHR were essential for promoting lipid metabolism and climbing behavior in response to HearNPV infection. Both molecular docking result and Ca2+ imaging showed that both HaAKH1 and HaAKH2 could interact with HaAKHR. Knockdown of HaAKH1, HaAKH2 and HaAKHR resulted in not only the accumulation of triacylglycerol (TAG), but also the reduction of the replication of HearNPV and the crawling ability of infected H. armigera larvae, resulting in a decrease in the final death height of the infected larvae. We further validated this conclusion by injecting active peptides of HaAKH1 and HaAKH2 to infected larvae. In addition, we investigated the downstream of HaAKH signaling and found that hormone-sensitive lipase (HaHSL) changed with changes in HaAKH signaling and HaHSL played the same role as HaAKH signaling. These findings not only revealed the mechanism by which parasites manipulated host lipid metabolism, but more significantly, explored the relationship between lipid metabolism and behavioral changes of hosts manipulated by parasites, broadening our understanding of the phenomenon of parasites manipulating host behavioral changes.
Collapse
Affiliation(s)
- Lin Zhu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yuqing Xie
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Chenxi Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Cheng
- Department of Entomology College of Plant Protection, Shanxi Agricultural University, Jinzhong, China
| | - Zhongjian Shen
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Key Laboratory of Natural Enemy Insects of Ministry of Agriculture and Rural Affairs, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xiaoming Liu
- State Key Laboratory of Wheat and Maize Crop Science, Henan International Laboratory for Green Pest Control, College of Plant Protection, Henan Agricultural University, Zhengzhou, China
| | - Limei Cai
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xinyuan Ning
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Songdou Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Zhen Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Qiuying Huang
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan, China
| | - Xiaoxia Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
da Silva GF, Rodrigues NR, Boligon AA, Ávila E, da Rosa Silva L, Franco JL, Posser T. Assessing the Effects of Palm Oil Consumption on Life Expectancy, Metabolic Markers, and Oxidative Stress in Drosophila melanogaster. Cell Biochem Funct 2024; 42:e70017. [PMID: 39567251 DOI: 10.1002/cbf.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/22/2024]
Abstract
Palm oil is the world's second most consumed vegetable oil, sourced from the tropical palm tree Elaeis guineensis. Its consumption has been associated with a higher incidence of cardiovascular disease, largely due to its elevated palmitic acid content, however those studies are contradictory and inconclusive. Wishing to contribute to this issue, the present study aims to investigate the molecular and toxicological effects of this oil and the involvement of oxidative stress, given its role in metabolic dysfunctions using Drosophila melanogaster. This study examines survival rates, and locomotor performance, oxidative status by analysis of lipid peroxidation, ROS formation, thiol levels and antioxidant enzyme activity, and metabolic parameters such as cholesterol and triglycerides, glucose, trehalose and glycogen levels. Exposure to palm oil concentrations of 10% and 30% resulted in a shortened lifespan, reduced locomotor performance, and increased lipid peroxidation, with lower thiol levels and antioxidant enzyme modulation. Cholesterol levels was increased whereas energetic fuels as glucose and glycogen and trehalose were decreased mainly after 10 days of exposure. These findings underscore the detrimental effects of high-fat diets containing palm oil on lifespan, antioxidant defenses, and metabolism in Drosophila melanogaster. This data highlights the potential risk associated with the habitual consumption of palm oil in the daily diet by population, particularly concerning cardiovascular health and metabolic function.
Collapse
Affiliation(s)
- Gustavo Felipe da Silva
- Oxidative Stress and Cell Signaling Research Group, Federal University of Pampa, Campus São Gabriel, São Gabriel, Rio Grande do Sul, Brazil
| | - Nathane Rosa Rodrigues
- Research Group of Biochemistry and Toxicology of Bioactive Compounds (GBToxBio), Federal University of Pampa, Campus Uruguaiana, Uruguaiana, Rio Grande do Sul, Brazil
| | - Aline Augusti Boligon
- Post-Graduation Program in Pharmaceutical Sciences, Federal University of Santa Maria, Santa Maria, Rio Grande do Sul, Brazil
| | - Eduardo Ávila
- Oxidative Stress and Cell Signaling Research Group, Federal University of Pampa, Campus São Gabriel, São Gabriel, Rio Grande do Sul, Brazil
| | - Luan da Rosa Silva
- Oxidative Stress and Cell Signaling Research Group, Federal University of Pampa, Campus São Gabriel, São Gabriel, Rio Grande do Sul, Brazil
| | - Jeferson Luis Franco
- Oxidative Stress and Cell Signaling Research Group, Federal University of Pampa, Campus São Gabriel, São Gabriel, Rio Grande do Sul, Brazil
| | - Thaís Posser
- Oxidative Stress and Cell Signaling Research Group, Federal University of Pampa, Campus São Gabriel, São Gabriel, Rio Grande do Sul, Brazil
| |
Collapse
|
16
|
Hunter‐Manseau F, Cormier SB, Strang R, Pichaud N. Fasting as a precursor to high-fat diet enhances mitochondrial resilience in Drosophila melanogaster. INSECT SCIENCE 2024; 31:1770-1788. [PMID: 38514255 PMCID: PMC11632299 DOI: 10.1111/1744-7917.13355] [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: 12/13/2023] [Revised: 01/29/2024] [Accepted: 02/08/2024] [Indexed: 03/23/2024]
Abstract
Changes in diet type and nutrient availability can impose significant environmental stress on organisms, potentially compromising physiological functions and reproductive success. In nature, dramatic fluctuations in dietary resources are often observed and adjustments to restore cellular homeostasis are crucial to survive this type of stress. In this study, we exposed male Drosophila melanogaster to two modulated dietary treatments: one without a fasting period before exposure to a high-fat diet and the other with a 24-h fasting period. We then investigated mitochondrial metabolism and molecular responses to these treatments. Exposure to a high-fat diet without a preceding fasting period resulted in disrupted mitochondrial respiration, notably at the level of complex I. On the other hand, a short fasting period before the high-fat diet maintained mitochondrial respiration. Generally, transcript abundance of genes associated with mitophagy, heat-shock proteins, mitochondrial biogenesis, and nutrient sensing pathways increased either slightly or significantly following a fasting period and remained stable when flies were subsequently put on a high-fat diet, whereas a drastic decrease of almost all transcript abundances was observed for all these pathways when flies were exposed directly to a high-fat diet. Moreover, mitochondrial enzymatic activities showed less variation after the fasting period than the treatment without a fasting period. Overall, our study sheds light on the mechanistic protective effects of fasting prior to a high-fat diet and highlights the metabolic flexibility of Drosophila mitochondria in response to abrupt dietary changes and have implication for adaptation of species to their changing environment.
Collapse
Affiliation(s)
- Florence Hunter‐Manseau
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| | - Simon B. Cormier
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| | - Rebekah Strang
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| | - Nicolas Pichaud
- Department of Chemistry and BiochemistryUniversité de MonctonMonctonNew BrunswickCanada
- New Brunswick Centre for Precision MedicineMonctonNew BrunswickCanada
| |
Collapse
|
17
|
Yuan Q, Luo M, Xie Y, Song W, Wang Y, Deng D, Chen S, Guo H. Chronic trans fatty acid consumption shortens lifespan in male Drosophila melanogaster on a high-sugar and high-fat diet. Biogerontology 2024; 25:1285-1297. [PMID: 38582786 DOI: 10.1007/s10522-024-10101-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 02/21/2024] [Indexed: 04/08/2024]
Abstract
Aging entails the progressive decline in the body's self-regulation and functionality over time. Notably, obesity and aging exhibit parallel phenotypes, with obesity further accelerating the aging process across multiple dimensions and diminishing lifespan. In this study, we explored the impact of trans fatty acid (TFA) consumption on the overall health and lifespan of male Drosophila melanogaster under an isocaloric high-sugar and high-fat diet. Our results indicate that TFA intake results in a shortened lifespan, elevated body weight, and increased triglyceride levels in flies fed a high-sugar and high-fat diet with equivalent caloric intake. Additionally, TFA exposure induces oxidative stress, locomotor deficits, and damage to the intestinal barrier in flies. Collectively, chronic TFA consumption expedites the aging process and reduces the lifespan of male Drosophila melanogaster. These results contribute supplementary evidence regarding the adverse health effects associated with TFAs.
Collapse
Affiliation(s)
- Qianhua Yuan
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Mengliu Luo
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Yutong Xie
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Wanhan Song
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Ya Wang
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Dazhang Deng
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Shuyan Chen
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China
| | - Honghui Guo
- Department of Nutrition, School of Public Health, Guangdong Medical University, Dongguan, 523808, China.
- Dongguan Key Laboratory of Prevention and Treatment of Chronic Noncommunicable Diseases, School of Public Health, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
18
|
Rundell TB, Baranski TJ. Insect Models to Study Human Lipid Metabolism Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 39405006 DOI: 10.1007/5584_2024_827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
Disorders of lipid metabolism such as obesity have become some of the most significant diseases of the twenty-first century. Despite these metabolic diseases affecting more than a third of the population in highly industrialized nations, the mechanisms underlying disease development remain poorly understood. Insect models, such as Drosophila melanogaster, offer a means of systematically examining conserved lipid metabolism and its pathology. Over the past several decades, Drosophila melanogaster has been used to greatly expand on our knowledge of metabolic disease, often taking advantage of the extensive genetic tools available to researchers. Additionally, Drosophila melanogaster has served and will continue to serve as a powerful tool for validating the results of genome-wide approaches to the study of diseases. This chapter explores the advancements of insect models in the study of lipid metabolism disorders as well as highlight opportunities for future areas of research.
Collapse
Affiliation(s)
- Thomas B Rundell
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Thomas J Baranski
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
19
|
Dos Santos E, Cochemé HM. How does a fly die? Insights into ageing from the pathophysiology of Drosophila mortality. GeroScience 2024; 46:4003-4015. [PMID: 38642259 PMCID: PMC11336040 DOI: 10.1007/s11357-024-01158-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/05/2024] [Indexed: 04/22/2024] Open
Abstract
The fruit fly Drosophila melanogaster is a common animal model in ageing research. Large populations of flies are used to study the impact of genetic, nutritional and pharmacological interventions on survival. However, the processes through which flies die and their relative prevalence in Drosophila populations are still comparatively unknown. Understanding the causes of death in an animal model is essential to dissect the lifespan-extending interventions that are organism- or disease-specific from those broadly applicable to ageing. Here, we review the pathophysiological processes that can lead to fly death and discuss their relation to ageing.
Collapse
Affiliation(s)
- Eliano Dos Santos
- MRC Laboratory of Medical Sciences (LMS), Hammersmith Hospital Campus, Du Cane Road, London, W12 0HS, UK
- Institute of Clinical Sciences, Hammersmith Hospital Campus, Imperial College London, Du Cane Road, London, W12 0HS, UK
| | - Helena M Cochemé
- MRC Laboratory of Medical Sciences (LMS), Hammersmith Hospital Campus, Du Cane Road, London, W12 0HS, UK.
- Institute of Clinical Sciences, Hammersmith Hospital Campus, Imperial College London, Du Cane Road, London, W12 0HS, UK.
| |
Collapse
|
20
|
Ma P, Zhang Y, Yin Y, Wang S, Chen S, Liang X, Li Z, Deng H. Gut microbiota metabolite tyramine ameliorates high-fat diet-induced insulin resistance via increased Ca 2+ signaling. EMBO J 2024; 43:3466-3493. [PMID: 38965418 PMCID: PMC11329785 DOI: 10.1038/s44318-024-00162-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
The gut microbiota and their metabolites are closely linked to obesity-related diseases, such as type 2 diabetes, but their causal relationship and underlying mechanisms remain largely elusive. Here, we found that dysbiosis-induced tyramine (TA) suppresses high-fat diet (HFD)-mediated insulin resistance in both Drosophila and mice. In Drosophila, HFD increases cytosolic Ca2+ signaling in enterocytes, which, in turn, suppresses intestinal lipid levels. 16 S rRNA sequencing and metabolomics revealed that HFD leads to increased prevalence of tyrosine decarboxylase (Tdc)-expressing bacteria and resulting tyramine production. Tyramine acts on the tyramine receptor, TyrR1, to promote cytosolic Ca2+ signaling and activation of the CRTC-CREB complex to transcriptionally suppress dietary lipid digestion and lipogenesis in enterocytes, while promoting mitochondrial biogenesis. Furthermore, the tyramine-induced cytosolic Ca2+ signaling is sufficient to suppress HFD-induced obesity and insulin resistance in Drosophila. In mice, tyramine intake also improves glucose tolerance and insulin sensitivity under HFD. These results indicate that dysbiosis-induced tyramine suppresses insulin resistance in both flies and mice under HFD, suggesting a potential therapeutic strategy for related metabolic disorders, such as diabetes.
Collapse
Affiliation(s)
- Peng Ma
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China
| | - Yao Zhang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China
| | - Youjie Yin
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China
| | - Saifei Wang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China
| | - Shuxin Chen
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China
| | - Xueping Liang
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China
| | - Zhifang Li
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China
| | - Hansong Deng
- Yangzhi Rehabilitation Hospital, Sunshine Rehabilitation Center, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 20092, Shanghai, China.
| |
Collapse
|
21
|
Bauer I, Rimbach G, Cordeiro S, Bosy-Westphal A, Weghuber J, Ipharraguerre IR, Lüersen K. A comprehensive in-vitro/ in-vivo screening toolbox for the elucidation of glucose homeostasis modulating properties of plant extracts (from roots) and its bioactives. Front Pharmacol 2024; 15:1396292. [PMID: 38989154 PMCID: PMC11233739 DOI: 10.3389/fphar.2024.1396292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/10/2024] [Indexed: 07/12/2024] Open
Abstract
Plant extracts are increasingly recognized for their potential in modulating (postprandial) blood glucose levels. In this context, root extracts are of particular interest due to their high concentrations and often unique spectrum of plant bioactives. To identify new plant species with potential glucose-lowering activity, simple and robust methodologies are often required. For this narrative review, literature was sourced from scientific databases (primarily PubMed) in the period from June 2022 to January 2024. The regulatory targets of glucose homeostasis that could be modulated by bioactive plant compounds were used as search terms, either alone or in combination with the keyword "root extract". As a result, we present a comprehensive methodological toolbox for studying the glucose homeostasis modulating properties of plant extracts and its constituents. The described assays encompass in-vitro investigations involving enzyme inhibition (α-amylase, α-glucosidase, dipeptidyl peptidase 4), assessment of sodium-dependent glucose transporter 1 activity, and evaluation of glucose transporter 4 translocation. Furthermore, we describe a patch-clamp technique to assess the impact of extracts on KATP channels. While validating in-vitro findings in living organisms is imperative, we introduce two screenable in-vivo models (the hen's egg test and Drosophila melanogaster). Given that evaluation of the bioactivity of plant extracts in rodents and humans represents the current gold standard, we include approaches addressing this aspect. In summary, this review offers a systematic guide for screening plant extracts regarding their influence on key regulatory elements of glucose homeostasis, culminating in the assessment of their potential efficacy in-vivo. Moreover, application of the presented toolbox might contribute to further close the knowledge gap on the precise mechanisms of action of plant-derived compounds.
Collapse
Affiliation(s)
- Ilka Bauer
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Gerald Rimbach
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Sönke Cordeiro
- Institute of Physiology, University of Kiel, Kiel, Germany
| | - Anja Bosy-Westphal
- Division of Human Nutrition, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Julian Weghuber
- Center of Excellence Food Technology and Nutrition, University of Applied Sciences Upper Austria, Wels, Austria
- FFoQSI—Austrian Competence Centre for Feed and Food Quality, Safety & Innovation, Tulln, Austria
| | - Ignacio R. Ipharraguerre
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| | - Kai Lüersen
- Division of Food Sciences, Institute of Human Nutrition and Food Science, University of Kiel, Kiel, Germany
| |
Collapse
|
22
|
Melkani Y, Pant A, Guo Y, Melkani GC. Automated assessment of cardiac dynamics in aging and dilated cardiomyopathy Drosophila models using machine learning. Commun Biol 2024; 7:702. [PMID: 38849449 PMCID: PMC11161577 DOI: 10.1038/s42003-024-06371-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
The Drosophila model is pivotal in deciphering the pathophysiological underpinnings of various human ailments, notably aging and cardiovascular diseases. Cutting-edge imaging techniques and physiology yield vast high-resolution videos, demanding advanced analysis methods. Our platform leverages deep learning to segment optical microscopy images of Drosophila hearts, enabling the quantification of cardiac parameters in aging and dilated cardiomyopathy (DCM). Validation using experimental datasets confirms the efficacy of our aging model. We employ two innovative approaches deep-learning video classification and machine-learning based on cardiac parameters to predict fly aging, achieving accuracies of 83.3% (AUC 0.90) and 79.1%, (AUC 0.87) respectively. Moreover, we extend our deep-learning methodology to assess cardiac dysfunction associated with the knock-down of oxoglutarate dehydrogenase (OGDH), revealing its potential in studying DCM. This versatile approach promises accelerated cardiac assays for modeling various human diseases in Drosophila and holds promise for application in animal and human cardiac physiology under diverse conditions.
Collapse
Affiliation(s)
- Yash Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Engineering Physics Department, College of Engineering, University of California, Berkeley, CA, USA
| | - Aniket Pant
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Materials Science and Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Yiming Guo
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Girish C Melkani
- Department of Pathology, Division of Molecular and Cellular Pathology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
23
|
Ouyang X, Matt A, Wang F, Gracheva E, Migunova E, Rajamani S, Dubrovsky EB, Zhou C. Attention LSTM U-Net model for Drosophila melanogaster heart tube segmentation in optical coherence microscopy images. BIOMEDICAL OPTICS EXPRESS 2024; 15:3639-3653. [PMID: 38867790 PMCID: PMC11166423 DOI: 10.1364/boe.523364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/22/2024] [Accepted: 04/28/2024] [Indexed: 06/14/2024]
Abstract
Optical coherence microscopy (OCM) imaging of the Drosophila melanogaster (fruit fly) heart tube has enabled the non-invasive characterization of fly heart physiology in vivo. OCM generates large volumes of data, making it necessary to automate image analysis. Deep-learning-based neural network models have been developed to improve the efficiency of fly heart image segmentation. However, image artifacts caused by sample motion or reflections reduce the accuracy of the analysis. To improve the precision and efficiency of image data analysis, we developed an Attention LSTM U-Net model (FlyNet3.0), which incorporates an attention learning mechanism to track the beating fly heart in OCM images. The new model has improved the intersection over union (IOU) compared to FlyNet2.0 + with reflection artifacts from 86% to 89% and with movement from 81% to 89%. We also extended the capabilities of OCM analysis through the introduction of an automated, in vivo heart wall thickness measurement method, which has been validated on a Drosophila model of cardiac hypertrophy. This work will enable the comprehensive, non-invasive characterization of fly heart physiology in a high-throughput manner.
Collapse
Affiliation(s)
- Xiangping Ouyang
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Abigail Matt
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Fei Wang
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Elena Gracheva
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Ekaterina Migunova
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | - Saathvika Rajamani
- Department of Biological Sciences, Fordham University, Bronx, NY 10458, USA
| | | | - Chao Zhou
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
24
|
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
|
25
|
Musselman LP, Truong HG, DiAngelo JR. Transcriptional Control of Lipid Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024. [PMID: 38782870 DOI: 10.1007/5584_2024_808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Transcriptional control of lipid metabolism uses a framework that parallels the control of lipid metabolism at the protein or enzyme level, via feedback and feed-forward mechanisms. Increasing the substrates for an enzyme often increases enzyme gene expression, for example. A paucity of product can likewise potentiate transcription or stability of the mRNA encoding the enzyme or enzymes needed to produce it. In addition, changes in second messengers or cellular energy charge can act as on/off switches for transcriptional regulators to control transcript (and protein) abundance. Insects use a wide range of DNA-binding transcription factors (TFs) that sense changes in the cell and its environment to produce the appropriate change in transcription at gene promoters. These TFs work together with histones, spliceosomes, and additional RNA processing factors to ultimately regulate lipid metabolism. In this chapter, we will first focus on the important TFs that control lipid metabolism in insects. Next, we will describe non-TF regulators of insect lipid metabolism such as enzymes that modify acetylation and methylation status, transcriptional coactivators, splicing factors, and microRNAs. To conclude, we consider future goals for studying the mechanisms underlying the control of lipid metabolism in insects.
Collapse
Affiliation(s)
- Laura Palanker Musselman
- Department of Biological Sciences, Binghamton University, State University of New York, Binghamton, NY, USA
| | - Huy G Truong
- Division of Science, Pennsylvania State University, Berks Campus, Reading, PA, USA
| | - Justin R DiAngelo
- Division of Science, Pennsylvania State University, Berks Campus, Reading, PA, USA.
| |
Collapse
|
26
|
Singh A, Abhilasha KV, Acharya KR, Liu H, Nirala NK, Parthibane V, Kunduri G, Abimannan T, Tantalla J, Zhu LJ, Acharya JK, Acharya UR. A nutrient responsive lipase mediates gut-brain communication to regulate insulin secretion in Drosophila. Nat Commun 2024; 15:4410. [PMID: 38782979 PMCID: PMC11116528 DOI: 10.1038/s41467-024-48851-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Pancreatic β cells secrete insulin in response to glucose elevation to maintain glucose homeostasis. A complex network of inter-organ communication operates to modulate insulin secretion and regulate glucose levels after a meal. Lipids obtained from diet or generated intracellularly are known to amplify glucose-stimulated insulin secretion, however, the underlying mechanisms are not completely understood. Here, we show that a Drosophila secretory lipase, Vaha (CG8093), is synthesized in the midgut and moves to the brain where it concentrates in the insulin-producing cells in a process requiring Lipid Transfer Particle, a lipoprotein originating in the fat body. In response to dietary fat, Vaha stimulates insulin-like peptide release (ILP), and Vaha deficiency results in reduced circulatory ILP and diabetic features including hyperglycemia and hyperlipidemia. Our findings suggest Vaha functions as a diacylglycerol lipase physiologically, by being a molecular link between dietary fat and lipid amplified insulin secretion in a gut-brain axis.
Collapse
Affiliation(s)
- Alka Singh
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | | | - Kathya R Acharya
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
- University of Cincinnati College of Medicine, 3230 Eden Ave, Cincinnati, OH, 45267, USA
| | - Haibo Liu
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Niraj K Nirala
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Velayoudame Parthibane
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Govind Kunduri
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Thiruvaimozhi Abimannan
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Jacob Tantalla
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Jairaj K Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA.
| | - Usha R Acharya
- Cancer and Developmental Biology Laboratory, National Cancer Institute, Frederick, MD, 21702, USA.
| |
Collapse
|
27
|
Mora I, Puiggròs F, Serras F, Gil-Cardoso K, Escoté X. Emerging models for studying adipose tissue metabolism. Biochem Pharmacol 2024; 223:116123. [PMID: 38484851 DOI: 10.1016/j.bcp.2024.116123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024]
Abstract
Understanding adipose metabolism is essential for addressing obesity and related health concerns. However, the ethical and scientific pressure to animal testing, aligning with the 3Rs, has triggered the implementation of diverse alternative models for analysing anomalies in adipose metabolism. In this review, we will address this issue from various perspectives. Traditional adipocyte cell cultures, whether animal or human-derived, offer a fundamental starting point. These systems have their merits but may not fully replicate in vivo complexity. Established cell lines are valuable for high-throughput screening but may lack the authenticity of primary-derived adipocytes, which closely mimic native tissue. To enhance model sophistication, spheroids have been introduced. These three-dimensional cultures better mimicking the in vivo microenvironment, enabling the study of intricate cell-cell interactions, gene expression, and metabolic pathways. Organ-on-a-chip (OoC) platforms take this further by integrating multiple cell types into microfluidic devices, simulating tissue-level functions. Adipose-OoC (AOoC) provides dynamic environments with applications spanning drug testing to personalized medicine and nutrition. Beyond in vitro models, genetically amenable organisms (Caenorhabditis elegans, Drosophila melanogaster, and zebrafish larvae) have become powerful tools for investigating fundamental molecular mechanisms that govern adipose tissue functions. Their genetic tractability allows for efficient manipulation and high-throughput studies. In conclusion, a diverse array of research models is crucial for deciphering adipose metabolism. By leveraging traditional adipocyte cell cultures, primary-derived cells, spheroids, AOoCs, and lower organism models, we bridge the gap between animal testing and a more ethical, scientifically robust, and human-relevant approach, advancing our understanding of adipose tissue metabolism and its impact on health.
Collapse
Affiliation(s)
- Ignasi Mora
- Brudy Technology S.L., 08006 Barcelona, Spain
| | - Francesc Puiggròs
- Eurecat, Centre Tecnològic de Catalunya, Biotechnology Area, 43204 Reus, Spain
| | - Florenci Serras
- Department of Genetics, Microbiology and Statistics, School of Biology, University of Barcelona and Institute of Biomedicine of the University of Barcelona, Diagonal 643, 08028 Barcelona, Spain
| | - Katherine Gil-Cardoso
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health Unit, 43204 Reus, Spain
| | - Xavier Escoté
- Eurecat, Centre Tecnològic de Catalunya, Nutrition and Health Unit, 43204 Reus, Spain.
| |
Collapse
|
28
|
Wang J, Gu J, Yi J, Li J, Li W, Zhai Z. High-fat diets induce inflammatory IMD/NFκB signaling via gut microbiota remodeling in Drosophila. Front Cell Infect Microbiol 2024; 14:1347716. [PMID: 38716198 PMCID: PMC11074423 DOI: 10.3389/fcimb.2024.1347716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/02/2024] [Indexed: 06/05/2024] Open
Abstract
High-fat diets (HFDs), a prevailing daily dietary style worldwide, induce chronic low-grade inflammation in the central nervous system and peripheral tissues, promoting a variety of diseases including pathologies associated with neuroinflammation. However, the mechanisms linking HFDs to inflammation are not entirely clear. Here, using a Drosophila HFD model, we explored the mechanism of HFD-induced inflammation in remote tissues. We found that HFDs activated the IMD/NFκB immune pathway in the head through remodeling of the commensal gut bacteria. Removal of gut microbiota abolished such HFD-induced remote inflammatory response. Further experiments revealed that HFDs significantly increased the abundance of Acetobacter malorum in the gut, and the re-association of this bacterium was sufficient to elicit inflammatory response in remote tissues. Mechanistically, Acetobacter malorum produced a greater amount of peptidoglycan (PGN), a well-defined microbial molecular pattern that enters the circulation and remotely activates an inflammatory response. Our results thus show that HFDs trigger inflammation mediated by a bacterial molecular pattern that elicits host immune response.
Collapse
Affiliation(s)
| | | | | | | | | | - Zongzhao Zhai
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, China
| |
Collapse
|
29
|
Dark C, Ali N, Golenkina S, Dhyani V, Blazev R, Parker BL, Murphy KT, Lynch GS, Senapati T, Millard SS, Judge SM, Judge AR, Giri L, Russell SM, Cheng LY. Mitochondrial fusion and altered beta-oxidation drive muscle wasting in a Drosophila cachexia model. EMBO Rep 2024; 25:1835-1858. [PMID: 38429578 PMCID: PMC11014992 DOI: 10.1038/s44319-024-00102-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/28/2024] [Accepted: 02/08/2024] [Indexed: 03/03/2024] Open
Abstract
Cancer cachexia is a tumour-induced wasting syndrome, characterised by extreme loss of skeletal muscle. Defective mitochondria can contribute to muscle wasting; however, the underlying mechanisms remain unclear. Using a Drosophila larval model of cancer cachexia, we observed enlarged and dysfunctional muscle mitochondria. Morphological changes were accompanied by upregulation of beta-oxidation proteins and depletion of muscle glycogen and lipid stores. Muscle lipid stores were also decreased in Colon-26 adenocarcinoma mouse muscle samples, and expression of the beta-oxidation gene CPT1A was negatively associated with muscle quality in cachectic patients. Mechanistically, mitochondrial defects result from reduced muscle insulin signalling, downstream of tumour-secreted insulin growth factor binding protein (IGFBP) homologue ImpL2. Strikingly, muscle-specific inhibition of Forkhead box O (FOXO), mitochondrial fusion, or beta-oxidation in tumour-bearing animals preserved muscle integrity. Finally, dietary supplementation with nicotinamide or lipids, improved muscle health in tumour-bearing animals. Overall, our work demonstrates that muscle FOXO, mitochondria dynamics/beta-oxidation and lipid utilisation are key regulators of muscle wasting in cancer cachexia.
Collapse
Affiliation(s)
- Callum Dark
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Nashia Ali
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sofya Golenkina
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Vaibhav Dhyani
- Bioimaging and Data Analysis Lab, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
- Optical Science Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Melbourne, VIC, Australia
| | - Ronnie Blazev
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Benjamin L Parker
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Kate T Murphy
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Tarosi Senapati
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Queensland, QLD, 4072, Australia
| | - S Sean Millard
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Queensland, QLD, 4072, Australia
| | - Sarah M Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Florida, FL, 32603, USA
| | - Andrew R Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Florida, FL, 32603, USA
| | - Lopamudra Giri
- Bioimaging and Data Analysis Lab, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Sarah M Russell
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Optical Science Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Melbourne, VIC, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
30
|
DeLoriea J, Millet-Boureima C, Gamberi C. Protocol to build a drug-testing pipeline using large populations of Drosophila melanogaster. STAR Protoc 2023; 4:102747. [PMID: 38103197 PMCID: PMC10751569 DOI: 10.1016/j.xpro.2023.102747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/05/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
As a small animal that recapitulates many fundamental aspects of human disease, Drosophila lends itself to probing the biological activity of molecules and drug candidates. Here, we present a protocol to build a drug-testing pipeline in Drosophila. We describe steps for generating synchronous populations of Bicaudal C mutants by genetic crossing and wild-type fly culturing for controlled compound administration and exemplary phenotypic assays. For complete details on the use and execution of this protocol, please refer to Millet-Boureima et al.,1 Millet-Boureima et al.,2 and Gamberi et al.3.
Collapse
Affiliation(s)
- Jay DeLoriea
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA
| | | | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29526, USA.
| |
Collapse
|
31
|
Hou WQ, Wen DT, Zhong Q, Mo L, Wang S, Yin XY, Ma XF. Physical exercise ameliorates age-related deterioration of skeletal muscle and mortality by activating Pten-related pathways in Drosophila on a high-salt diet. FASEB J 2023; 37:e23304. [PMID: 37971426 DOI: 10.1096/fj.202301099r] [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: 06/02/2023] [Revised: 09/24/2023] [Accepted: 10/26/2023] [Indexed: 11/19/2023]
Abstract
The phosphatase and tensin congeners (Pten) gene affects cell growth, cell proliferation, and rearrangement of connections, and it is closely related to cellular senescence, but it remains unclear the role of muscle-Pten gene in exercise against age-related deterioration in skeletal muscle and mortality induced by a high-salt diet (HSD). In here, overexpression and knockdown of muscle Pten gene were constructed by building MhcGAL4 /PtenUAS-overexpression and MhcGAL4 /PtenUAS-RNAi system in flies, and flies were given exercise training and a HSD for 2 weeks. The results showed that muscle Pten knockdown significantly reduced the climbing speed, climbing endurance, GPX activity, and the expression of Pten, Sirt1, PGC-1α genes, and it significantly increased the expression of Akt and ROS level, and impaired myofibril and mitochondria of aged skeletal muscle. Pten knockdown prevented exercise from countering the HSD-induced age-related deterioration of skeletal muscle. Pten overexpression has the opposite effect on skeletal muscle aging when compared to it knockdown, and it promoted exercise against HSD-induced age-related deterioration of skeletal muscle. Pten overexpression significantly increased lifespan, but its knockdown significantly decreased lifespan of flies. Thus, current results confirmed that differential expression of muscle Pten gene played an important role in regulating skeletal muscle aging and lifespan, and it also affected the adaptability of aging skeletal muscle to physical exercise since it determined the activity of muscle Pten/Akt pathway and Pten/Sirt1/PGC-1α pathway.
Collapse
Affiliation(s)
- Wen-Qi Hou
- Department of Physical Education, Ludong University, Yantai, China
| | - Deng-Tai Wen
- Department of Physical Education, Ludong University, Yantai, China
| | - Qi Zhong
- Department of Physical Education, Ludong University, Yantai, China
| | - Lan Mo
- Department of Physical Education, Hainan Normal University, Haikou, China
| | - Shuo Wang
- Department of Physical Education, Ludong University, Yantai, China
| | - Xin-Yuan Yin
- Department of Physical Education, Ludong University, Yantai, China
| | - Xing-Feng Ma
- Department of Physical Education, Ludong University, Yantai, China
| |
Collapse
|
32
|
Bosch JA, Keith N, Escobedo F, Fisher WW, LaGraff JT, Rabasco J, Wan KH, Weiszmann R, Hu Y, Kondo S, Brown JB, Perrimon N, Celniker SE. Molecular and functional characterization of the Drosophila melanogaster conserved smORFome. Cell Rep 2023; 42:113311. [PMID: 37889754 PMCID: PMC10843857 DOI: 10.1016/j.celrep.2023.113311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/24/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Short polypeptides encoded by small open reading frames (smORFs) are ubiquitously found in eukaryotic genomes and are important regulators of physiology, development, and mitochondrial processes. Here, we focus on a subset of 298 smORFs that are evolutionarily conserved between Drosophila melanogaster and humans. Many of these smORFs are conserved broadly in the bilaterian lineage, and ∼182 are conserved in plants. We observe remarkably heterogeneous spatial and temporal expression patterns of smORF transcripts-indicating wide-spread tissue-specific and stage-specific mitochondrial architectures. In addition, an analysis of annotated functional domains reveals a predicted enrichment of smORF polypeptides localizing to mitochondria. We conduct an embryonic ribosome profiling experiment and find support for translation of 137 of these smORFs during embryogenesis. We further embark on functional characterization using CRISPR knockout/activation, RNAi knockdown, and cDNA overexpression, revealing diverse phenotypes. This study underscores the importance of identifying smORF function in disease and phenotypic diversity.
Collapse
Affiliation(s)
- Justin A Bosch
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Nathan Keith
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Felipe Escobedo
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - William W Fisher
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - James Thai LaGraff
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Jorden Rabasco
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kenneth H Wan
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Richard Weiszmann
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Shu Kondo
- Laboratory of Invertebrate Genetics, National Institute of Genetics, Mishima, Shizuoka 411-8540, Japan
| | - James B Brown
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Division of Environmental Genomics and Systems Biology, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Susan E Celniker
- Division of Biological Systems and Engineering, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
33
|
Mirzoyan Z, Valenza A, Zola S, Bonfanti C, Arnaboldi L, Ferrari N, Pollard J, Lupi V, Cassinelli M, Frattaroli M, Sahin M, Pasini ME, Bellosta P. A Drosophila model targets Eiger/TNFα to alleviate obesity-related insulin resistance and macrophage infiltration. Dis Model Mech 2023; 16:dmm050388. [PMID: 37828911 PMCID: PMC10651092 DOI: 10.1242/dmm.050388] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/05/2023] [Indexed: 10/14/2023] Open
Abstract
Obesity is associated with various metabolic disorders, such as insulin resistance and adipose tissue inflammation (ATM), characterized by macrophage infiltration into adipose cells. This study presents a new Drosophila model to investigate the mechanisms underlying these obesity-related pathologies. We employed genetic manipulation to reduce ecdysone levels to prolong the larval stage. These animals are hyperphagic and exhibit features resembling obesity in mammals, including increased lipid storage, adipocyte hypertrophy and high circulating glucose levels. Moreover, we observed significant infiltration of immune cells (hemocytes) into the fat bodies, accompanied by insulin resistance. We found that attenuation of Eiger/TNFα signaling reduced ATM and improved insulin sensitivity. Furthermore, using metformin and the antioxidants anthocyanins, we ameliorated both phenotypes. Our data highlight evolutionarily conserved mechanisms allowing the development of Drosophila models for discovering therapeutic pathways in adipose tissue immune cell infiltration and insulin resistance. Our model can also provide a platform to perform genetic screens or test the efficacy of therapeutic interventions for diseases such as obesity, type 2 diabetes and non-alcoholic fatty liver disease.
Collapse
Affiliation(s)
- Zhasmine Mirzoyan
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Alice Valenza
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Sheri Zola
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - Carola Bonfanti
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | | | - Nicholas Ferrari
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
| | - John Pollard
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | - Valeria Lupi
- Department of Biosciences, University of Milan, 20133 Milan, Italy
| | | | | | - Mehtap Sahin
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
- Department of Biology, University of Ankara, 06110 Ankara, Turkey
| | | | - Paola Bellosta
- Department of Computational, Cellular and Integrative Biology (CIBIO), University of Trento, 38123 Trento, Italy
- Department of Medicine, NYU Langone Medical Center, 10016 New York, USA
| |
Collapse
|
34
|
Wen D, Chen Y, Tian X, Hou W. Physical exercise improves the premature muscle aging and lifespan reduction induced by high-salt intake and muscle CG2196(salt) overexpression in Drosophila. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
35
|
Yan H, Ding M, Peng T, Zhang P, Tian R, Zheng L. Regular Exercise Modulates the dfoxo/ dsrebp Pathway to Alleviate High-Fat-Diet-Induced Obesity and Cardiac Dysfunction in Drosophila. Int J Mol Sci 2023; 24:15562. [PMID: 37958546 PMCID: PMC10650635 DOI: 10.3390/ijms242115562] [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: 09/11/2023] [Revised: 10/22/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023] Open
Abstract
Obesity is a prevalent metabolic disorder associated with various diseases, including cardiovascular conditions. While exercise is recognized as an effective approach for preventing and treating obesity, its underlying molecular mechanisms remain unclear. This study aimed to explore the impact of regular exercise on high-fat-diet-induced obesity and cardiac dysfunction in Drosophila, shedding light on its molecular mechanisms by identifying its regulation of the dfoxo and dsrebp signaling pathways. Our findings demonstrated that a high-fat diet leads to weight gain, fat accumulation, reduced climbing performance, and elevated triglyceride levels in Drosophila. Additionally, cardiac microfilaments in these flies exhibited irregularities, breakages, and shortening. M-mode analysis revealed that high-fat-diet-fed Drosophila displayed increased heart rates, shortened cardiac cycles, decreased systolic intervals, heightened arrhythmia indices, reduced diastolic diameters, and diminished fractional shortening. Remarkably, regular exercise effectively ameliorated these adverse outcomes. Further analysis showed that regular exercise reduced fat synthesis, promoted lipolysis, and mitigated high-fat-diet-induced cardiac dysfunction in Drosophila. These results suggest that regular exercise may mitigate high-fat-diet-induced obesity and cardiac dysfunction in Drosophila by regulating the dfoxo and dsrebp signaling pathways, offering valuable insights into the mechanisms underlying the beneficial effects of exercise on obesity and cardiac dysfunction induced by a high-fat diet.
Collapse
Affiliation(s)
| | | | | | | | | | - Lan Zheng
- Key Laboratory of Physical Fitness and Exercise Rehabilitation of Hunan Province, College of Physical Education, Hunan Normal University, Changsha 410012, China; (H.Y.); (M.D.); (T.P.); (P.Z.); (R.T.)
| |
Collapse
|
36
|
Yang J, Tang R, Chen S, Chen Y, Yuan K, Huang R, Wang L. Exposure to high-sugar diet induces transgenerational changes in sweet sensitivity and feeding behavior via H3K27me3 reprogramming. eLife 2023; 12:e85365. [PMID: 37698486 PMCID: PMC10558205 DOI: 10.7554/elife.85365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 09/11/2023] [Indexed: 09/13/2023] Open
Abstract
Human health is facing a host of new threats linked to unbalanced diets, including high-sugar diet (HSD), which contributes to the development of both metabolic and behavioral disorders. Studies have shown that diet-induced metabolic dysfunctions can be transmitted to multiple generations of offspring and exert long-lasting health burden. Meanwhile, whether and how diet-induced behavioral abnormalities can be transmitted to the offspring remains largely unclear. Here, we showed that ancestral HSD exposure suppressed sweet sensitivity and feeding behavior in the offspring in Drosophila. These behavioral deficits were transmitted through the maternal germline and companied by the enhancement of H3K27me3 modifications. PCL-PRC2 complex, a major driver of H3K27 trimethylation, was upregulated by ancestral HSD exposure, and disrupting its activity eliminated the transgenerational inheritance of sweet sensitivity and feeding behavior deficits. Elevated H3K27me3 inhibited the expression of a transcriptional factor Cad and suppressed sweet sensitivity of the sweet-sensing gustatory neurons, reshaping the sweet perception and feeding behavior of the offspring. Taken together, we uncovered a novel molecular mechanism underlying behavioral abnormalities spanning multiple generations of offspring upon ancestral HSD exposure, which would contribute to the further understanding of long-term health risk of unbalanced diet.
Collapse
Affiliation(s)
- Jie Yang
- Life Sciences Institute, Zhejiang UniversityHangzhouChina
| | - Ruijun Tang
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South UniversityChangshaChina
| | - Shiye Chen
- Life Sciences Institute, Zhejiang UniversityHangzhouChina
| | - Yinan Chen
- Life Sciences Institute, Zhejiang UniversityHangzhouChina
| | - Kai Yuan
- Hunan Key Laboratory of Molecular Precision Medicine, Department of Neurosurgery, Xiangya Hospital, and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South UniversityChangshaChina
- The Biobank of Xiangya Hospital, Xiangya Hospital, Central South UniversityChangshaChina
| | - Rui Huang
- Center for Neurointelligence, School of Medicine, Chongqing UniversityChongqingChina
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
| | - Liming Wang
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
| |
Collapse
|
37
|
Liu J, Zhang Y, Wang QQ, Zhou Y, Liu JL. Fat body-specific reduction of CTPS alleviates HFD-induced obesity. eLife 2023; 12:e85293. [PMID: 37695169 PMCID: PMC10495109 DOI: 10.7554/elife.85293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 08/25/2023] [Indexed: 09/12/2023] Open
Abstract
Obesity induced by high-fat diet (HFD) is a multi-factorial disease including genetic, physiological, behavioral, and environmental components. Drosophila has emerged as an effective metabolic disease model. Cytidine 5'-triphosphate synthase (CTPS) is an important enzyme for the de novo synthesis of CTP, governing the cellular level of CTP and the rate of phospholipid synthesis. CTPS is known to form filamentous structures called cytoophidia, which are found in bacteria, archaea, and eukaryotes. Our study demonstrates that CTPS is crucial in regulating body weight and starvation resistance in Drosophila by functioning in the fat body. HFD-induced obesity leads to increased transcription of CTPS and elongates cytoophidia in larval adipocytes. Depleting CTPS in the fat body prevented HFD-induced obesity, including body weight gain, adipocyte expansion, and lipid accumulation, by inhibiting the PI3K-Akt-SREBP axis. Furthermore, a dominant-negative form of CTPS also prevented adipocyte expansion and downregulated lipogenic genes. These findings not only establish a functional link between CTPS and lipid homeostasis but also highlight the potential role of CTPS manipulation in the treatment of HFD-induced obesity.
Collapse
Affiliation(s)
- Jingnan Liu
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- College of Life Sciences, Shanghai Normal UniversityShanghaiChina
| | - Yuanbing Zhang
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Qiao-Qi Wang
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Youfang Zhou
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of SciencesShanghaiChina
- University of Chinese Academy of SciencesBeijingChina
| | - Ji-Long Liu
- School of Life Science and Technology, ShanghaiTech UniversityShanghaiChina
- Department of Physiology, Anatomy and Genetics, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
38
|
Yagihashi S. Contribution of animal models to diabetes research: Its history, significance, and translation to humans. J Diabetes Investig 2023; 14:1015-1037. [PMID: 37401013 PMCID: PMC10445217 DOI: 10.1111/jdi.14034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/10/2023] [Accepted: 05/16/2023] [Indexed: 07/05/2023] Open
Abstract
Diabetes mellitus is still expanding globally and is epidemic in developing countries. The combat of this plague has caused enormous economic and social burdens related to a lowered quality of life in people with diabetes. Despite recent significant improvements of life expectancy in patients with diabetes, there is still a need for efforts to elucidate the complexities and mechanisms of the disease processes to overcome this difficult disorder. To this end, the use of appropriate animal models in diabetes studies is invaluable for translation to humans and for the development of effective treatment. In this review, a variety of animal models of diabetes with spontaneous onset in particular will be introduced and discussed for their implication in diabetes research.
Collapse
Affiliation(s)
- Soroku Yagihashi
- Department of Exploratory Medicine for Nature, Life and HumansToho University School of MedicineChibaJapan
- Department of PathologyHirosaki University Graduate School of MedicineHirosakiJapan
| |
Collapse
|
39
|
Ye L, Jiang X, Chen L, Chen S, Li H, Du R, You W, Peng J, Guo P, Zhang R, Yu H, Dong G, Li D, Li X, Chen W, Xing X, Xiao Y. Moderate body lipid accumulation in mice attenuated benzene-induced hematotoxicity via acceleration of benzene metabolism and clearance. ENVIRONMENT INTERNATIONAL 2023; 178:108113. [PMID: 37506515 DOI: 10.1016/j.envint.2023.108113] [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: 04/17/2023] [Revised: 06/16/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Recent population and animal studies have revealed a correlation between fat content and the severity of benzene-induced hematologic toxicity. However, the precise impact of lipid deposition on benzene-induced hematotoxicity and the underlying mechanisms remain unclear. In this study, we established a mouse model with moderate lipid accumulation by subjecting the mice to an 8-week high-fat diet (45% kcal from fat, HFD), followed by 28-day inhalation of benzene at doses of 0, 1, 10, and 100 ppm. The results showed that benzene exposure caused a dose-dependent reduction of peripheral white blood cell (WBC) counts in both diet groups. Notably, this reduction was less pronounced in the HFD-fed mice, suggesting that moderate lipid accumulation mitigates benzene-related hematotoxicity. To investigate the molecular basis for this effect, we performed bioinformatics analysis of high-throughput transcriptome sequencing data, which revealed that moderate lipid deposition alters mouse metabolism and stress tolerance towards xenobiotics. Consistently, the expression of key metabolic enzymes, such as Cyp2e1 and Gsta1, were upregulated in the HFD-fed mice upon benzene exposure. Furthermore, we utilized a real-time exhaled breath detection technique to monitor exhaled benzene metabolites, and the results indicated that moderate lipid deposition enhanced metabolic activation and increased the elimination of benzene metabolites. Collectively, these findings demonstrate that moderate lipid deposition confers reduced susceptibility to benzene-induced hematotoxicity in mice, at least in part, by accelerating benzene metabolism and clearance.
Collapse
Affiliation(s)
- Lizhu Ye
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xinhang Jiang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Liping Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Huiyao Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui Du
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei You
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Jing Peng
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ping Guo
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui Zhang
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Hongyao Yu
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Guanghui Dong
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Daochuan Li
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xue Li
- Institute of Mass Spectrometry and Atmospheric Environment, Jinan University, Guangzhou 510632, China
| | - Wen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiumei Xing
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| | - Yongmei Xiao
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
40
|
Mirzoyan Z, Valenza A, Zola S, Bonfanti C, Arnaboldi L, Ferrari N, Pollard J, Lupi V, Cassinelli M, Frattaroli M, Sahin M, Pasini ME, Bellosta P. A Novel Drosophila Model to Investigate Adipose Tissue Macrophage Infiltration (ATM) and Obesity highlights the Therapeutic Potential of Attenuating Eiger/TNFα Signaling to Ameliorate Insulin Resistance and ATM. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.06.548016. [PMID: 37461586 PMCID: PMC10350075 DOI: 10.1101/2023.07.06.548016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Obesity is a global health concern associated with various metabolic disorders including insulin resistance and adipose tissue inflammation characterized by adipose tissue macrophage (ATM) infiltration. In this study, we present a novel Drosophila model to investigate the mechanisms underlying ATM infiltration and its association with obesity-related pathologies. Furthermore, we demonstrate the therapeutic potential of attenuating Eiger/TNFα signaling to ameliorate insulin resistance and ATM. To study ATM infiltration and its consequences, we established a novel Drosophila model (OBL) that mimics key aspects of human adipose tissue and allows for investigating ATM infiltration and other related metabolic disorders in a controlled experimental system. We employed genetic manipulation to reduce ecdysone levels to prolong the larval stage. These animals are hyperphagic, and exhibit features resembling obesity in mammals, including increased lipid storage, adipocyte hypertrophy, and high levels of circulating glucose. Moreover, we observed a significant infiltration of immune cells (hemocytes) in the fat bodies accompanied by insulin resistance and systemic metabolic dysregulation. Furthermore, we found that attenuation of Eiger/TNFα signaling and using metformin and anti-oxidant bio-products like anthocyanins led to a reduction in ATM infiltration and improved insulin sensitivity. Our data suggest that the key mechanisms that trigger immune cell infiltration into adipose tissue are evolutionarily conserved and may provide the opportunity to develop Drosophila models to better understand pathways critical for immune cell recruitment into adipose tissue, in relation to the development of insulin resistance in metabolic diseases such as obesity and type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD). We believe that our OBL model can also be a valuable tool and provide a platform either to perform genetic screens or to test the efficacy and safety of novel therapeutic interventions for these diseases.
Collapse
|
41
|
Kervadec A, Kezos J, Ni H, Yu M, Marchant J, Spiering S, Kannan S, Kwon C, Andersen P, Bodmer R, Grandi E, Ocorr K, Colas AR. Multiplatform modeling of atrial fibrillation identifies phospholamban as a central regulator of cardiac rhythm. Dis Model Mech 2023; 16:dmm049962. [PMID: 37293707 PMCID: PMC10387351 DOI: 10.1242/dmm.049962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 05/26/2023] [Indexed: 06/10/2023] Open
Abstract
Atrial fibrillation (AF) is a common and genetically inheritable form of cardiac arrhythmia; however, it is currently not known how these genetic predispositions contribute to the initiation and/or maintenance of AF-associated phenotypes. One major barrier to progress is the lack of experimental systems to investigate the effects of gene function on rhythm parameters in models with human atrial and whole-organ relevance. Here, we assembled a multi-model platform enabling high-throughput characterization of the effects of gene function on action potential duration and rhythm parameters using human induced pluripotent stem cell-derived atrial-like cardiomyocytes and a Drosophila heart model, and validation of the findings using computational models of human adult atrial myocytes and tissue. As proof of concept, we screened 20 AF-associated genes and identified phospholamban loss of function as a top conserved hit that shortens action potential duration and increases the incidence of arrhythmia phenotypes upon stress. Mechanistically, our study reveals that phospholamban regulates rhythm homeostasis by functionally interacting with L-type Ca2+ channels and NCX. In summary, our study illustrates how a multi-model system approach paves the way for the discovery and molecular delineation of gene regulatory networks controlling atrial rhythm with application to AF.
Collapse
Affiliation(s)
- Anaïs Kervadec
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - James Kezos
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Haibo Ni
- Department of Pharmacology, UC Davis, Davis, CA 95616, USA
| | - Michael Yu
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - James Marchant
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Sean Spiering
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Suraj Kannan
- Johns Hopkins University, Baltimore, MD 21205, USA
| | - Chulan Kwon
- Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Rolf Bodmer
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | | | - Karen Ocorr
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Alexandre R. Colas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| |
Collapse
|
42
|
Asiimwe OH, Rubaihayo J, Sulaiman SO, Osuwat LO, Kasozi KI. A protein restricted diet induces a stable increased fat storage phenotype in flies. Toxicol Rep 2023; 10:706-713. [PMID: 37396850 PMCID: PMC10313861 DOI: 10.1016/j.toxrep.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 07/04/2023] Open
Abstract
Background Scientific evidence has revealed possible confounders in diet induced obesity models of Drosophila melanogaster. High Sugar Diet (HSD) induction of obesity in flies has been associated with fly hyperosmolarity and glucotoxicity, while High Fat Diet (HFD) induction has been associated with lipotoxicity. The objective of this study was to assess for a healthy obesity phenotype by comparison of fly survival, physio-chemical and biochemical changes associated with HSD, HFD and Protein Restricted Diet (PRD) obesity induction models of male Drosophila melanogaster. Here, we provide information on a PRD as the plausible option in obesity research not involving cancer, diabetes, glucotoxicity and lipotoxicity studies. Methods Obesity was induced by exposing Drosophila melanogaster white mutant w1118 to four experimental diets for four weeks. Group 1 was fed regular food (control), group 2 was fed a 0.5% less yeast than in regular feed (PRD), group 3 was fed a 30% w/v sucrose to regular cornmeal food (HSD) and group 4 was fed a 10% w/v food-grade coconut oil to regular cornmeal food (HFD). Peristaltic waves were measured on 3rd instar larvae of all experimental groups. Negative geotaxis, fly survival, body mass, catalase activity, triglycerides (TG/TP), sterol, and total protein were measured in adult Drosophila melanogaster after four weeks. Results Triglycerides (TG/TP) and total protein levels were significantly higher in HSD phenotype. Sterols were higher in HFD phenotype. Though catalase enzyme activity was highest in PRD phenotype, this activity was not statistically significant when compared to that of HSD and HFD phenotypes. However, PRD phenotype had the lowest mass, highest survival rate and the highest negative geotaxis, thus demonstrating a balanced, stable and more viable metabolic status in the experimental model. Conclusion A protein restricted diet induces a stable increased fat storage phenotype in Drosophila melanogaster.
Collapse
Affiliation(s)
- Oscar Hilary Asiimwe
- Faculty of Health Sciences, Mountains of the Moon University, Box 837, Fort Portal, Uganda
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, International Campus, Zhejiang University, Haining, China
| | - John Rubaihayo
- Faculty of Health Sciences, Mountains of the Moon University, Box 837, Fort Portal, Uganda
| | - Sheu Oluwadare Sulaiman
- Graduate Program in Cell Biology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
- Department of Physiology, School of Medicine and Pharmacy, College of Medicine and Health Sciences, University of Rwanda, Huye, Rwanda
| | | | - Keneth Iceland Kasozi
- School of Medicine, Kabale University, Box 317, Kabale, Uganda
- Infection Medicine, Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, EH8 9JZ, Edinburgh, United Kingdom
| |
Collapse
|
43
|
Zhao Y, van de Leemput J, Han Z. The opportunities and challenges of using Drosophila to model human cardiac diseases. Front Physiol 2023; 14:1182610. [PMID: 37123266 PMCID: PMC10130661 DOI: 10.3389/fphys.2023.1182610] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/05/2023] [Indexed: 05/02/2023] Open
Abstract
The Drosophila heart tube seems simple, yet it has notable anatomic complexity and contains highly specialized structures. In fact, the development of the fly heart tube much resembles that of the earliest stages of mammalian heart development, and the molecular-genetic mechanisms driving these processes are highly conserved between flies and humans. Combined with the fly's unmatched genetic tools and a wide variety of techniques to assay both structure and function in the living fly heart, these attributes have made Drosophila a valuable model system for studying human heart development and disease. This perspective focuses on the functional and physiological similarities between fly and human hearts. Further, it discusses current limitations in using the fly, as well as promising prospects to expand the capabilities of Drosophila as a research model for studying human cardiac diseases.
Collapse
Affiliation(s)
- Yunpo Zhao
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Joyce van de Leemput
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Zhe Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
44
|
Moura-Silva J, Tavares MPS, Almeida-Oliveira F, Majerowicz D. Diet supplementation with egg yolk powder fattens the beetle Tribolium castaneum. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2023; 112:e22000. [PMID: 36656770 DOI: 10.1002/arch.22000] [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: 05/18/2022] [Revised: 11/11/2022] [Accepted: 01/03/2023] [Indexed: 06/17/2023]
Abstract
Insects have become essential models in studying human metabolic diseases, mainly due to their low maintenance cost and available tools. Both mutations and modified diets induce metabolic states similar to human obesity and diabetes. Here, we explore the effect of a high-calorie, high-fat diet on the metabolism of the beetle Tribolium castaneum. Supplementation of the wheat flour diet with powdered egg yolk for 3 weeks increased the total triacylglycerol and accelerated larval development. In addition, this diet increased the triacylglycerol levels of adult beetles. However, this egg yolk supplementation did not alter the larvae's total glucose levels or lipogenic capacity and ATP citrate lyase activity. The diet also did not change the expression profile of several lipid and carbohydrate metabolism genes and insulin-like peptides. Thus, we conclude that the diet supplemented with egg yolk induces increased fat without causing diabetes phenotypes, as seen in other hypercaloric diets in insects.
Collapse
Affiliation(s)
- Julia Moura-Silva
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Matheus P S Tavares
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - David Majerowicz
- Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular - INCT-EM, Rio de Janeiro, Brazil
| |
Collapse
|
45
|
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
|
46
|
Rezaie J, Aboulhassani A, Keyhanmanesh R, Rahbarghazi R, Delkhosh A, Salimi L, Zamani AN, Rahbarghazi A, Ahmadi M, Ghiasi F. Effect of voluntary wheel running on autophagy status in lung tissue of high-fat diet-fed rats. COMPARATIVE EXERCISE PHYSIOLOGY 2023. [DOI: 10.3920/cep210022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
Abstract
Here, we aimed to explore the therapeutic effect of voluntary wheel running (VWR) in high-fat diet-fed rats on pulmonary tissue injury via the modulation of autophagic response. Thirty-two rats were allocated into four groups; normal diet (Control); VWR; high-fat-diet (HFD), and HFD + VWR. After three months, pathological effect of HFD on pulmonary tissue was investigated. The levels of tumour necrosis factor (TNF)-α were detected in the bronchoalveolar lavage fluid (BALF). We monitored the expression of interleukin (IL)-6 and autophagy-related genes in lung tissues. H&E staining showed pathological changes in HFD group coincided with the increase of TNF-α levels in the bronchoalveolar fluid compared to the normal rats. Our results showed the up-regulation of IL-6, becline-1, LC3 and P62 in the HFD group compared to the Control group. VWR inhibited HFD-induced changes and could decrease HFD-induced changes via the regulation of autophagy status.
Collapse
Affiliation(s)
- J. Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, 5714783734 Urmia, Iran
| | - A. Aboulhassani
- Student Research Committee, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
- Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
| | - R. Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
| | - R. Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
| | - A. Delkhosh
- Stem Cell Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
| | - L. Salimi
- Stem Cell Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
| | - A.R. Nezhad Zamani
- Stem Cell Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
| | - A. Rahbarghazi
- Department of Physical Education and Sports Sciences, Faculty of Educational Science and Psychology, University of Mohaghegh Ardabil, 56199-11367 Ardabil, Iran
| | - M. Ahmadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, 5165665931 Tabriz, Iran
| | - F. Ghiasi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
47
|
Ren J, Zeng Q, Wu H, Liu X, Guida MC, Huang W, Zhai Y, Li J, Ocorr K, Bodmer R, Tang M. Deacetylase-dependent and -independent role of HDAC3 in cardiomyopathy. J Cell Physiol 2023; 238:647-658. [PMID: 36745702 PMCID: PMC10152801 DOI: 10.1002/jcp.30957] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 12/14/2022] [Accepted: 01/11/2023] [Indexed: 02/08/2023]
Abstract
Cardiomyopathy is a common disease of cardiac muscle that negatively affects cardiac function. HDAC3 commonly functions as corepressor by removing acetyl moieties from histone tails. However, a deacetylase-independent role of HDAC3 has also been described. Cardiac deletion of HDAC3 causes reduced cardiac contractility accompanied by lipid accumulation, but the molecular function of HDAC3 in cardiomyopathy remains unknown. We have used powerful genetic tools in Drosophila to investigate the enzymatic and nonenzymatic roles of HDAC3 in cardiomyopathy. Using the Drosophila heart model, we showed that cardiac-specific HDAC3 knockdown (KD) leads to prolonged systoles and reduced cardiac contractility. Immunohistochemistry revealed structural abnormalities characterized by myofiber disruption in HDAC3 KD hearts. Cardiac-specific HDAC3 KD showed increased levels of whole-body triglycerides and increased fibrosis. The introduction of deacetylase-dead HDAC3 mutant in HDAC3 KD background showed comparable results with wild-type HDAC3 in aspects of contractility and Pericardin deposition. However, deacetylase-dead HDAC3 mutants failed to improve triglyceride accumulation. Our data indicate that HDAC3 plays a deacetylase-independent role in maintaining cardiac contractility and preventing Pericardin deposition as well as a deacetylase-dependent role to maintain triglyceride homeostasis.
Collapse
Affiliation(s)
- Jieyu Ren
- Department of Biochemistry and Molecular Biology, College of Hengyang Medical, University of South China, Hengyang, China
| | - Qun Zeng
- Department of Biochemistry and Molecular Biology, College of Hengyang Medical, University of South China, Hengyang, China
| | - Hongmei Wu
- Department of Biochemistry and Molecular Biology, College of Hengyang Medical, University of South China, Hengyang, China
| | - Xuewen Liu
- Department of Biochemistry and Molecular Biology, College of Hengyang Medical, University of South China, Hengyang, China
| | - Maria C. Guida
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Wen Huang
- Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, China
| | - Yiyuan Zhai
- Department of Biochemistry and Molecular Biology, College of Hengyang Medical, University of South China, Hengyang, China
| | - Junjie Li
- Department of Biochemistry and Molecular Biology, College of Hengyang Medical, University of South China, Hengyang, China
| | - Karen Ocorr
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Rolf Bodmer
- Development Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Min Tang
- Department of Biochemistry and Molecular Biology, College of Hengyang Medical, University of South China, Hengyang, China
| |
Collapse
|
48
|
Regular Exercise in Drosophila Prevents Age-Related Cardiac Dysfunction Caused by High Fat and Heart-Specific Knockdown of skd. Int J Mol Sci 2023; 24:ijms24021216. [PMID: 36674733 PMCID: PMC9865808 DOI: 10.3390/ijms24021216] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Skuld (skd) is a subunit of the Mediator complex subunit complex. In the heart, skd controls systemic obesity, is involved in systemic energy metabolism, and is closely linked to cardiac function and aging. However, it is unclear whether the effect of cardiac skd on cardiac energy metabolism affects cardiac function. We found that cardiac-specific knockdown of skd showed impaired cardiac function, metabolic impairment, and premature aging. Drosophila was subjected to an exercise and high-fat diet (HFD) intervention to explore the effects of exercise on cardiac skd expression and cardiac function in HFD Drosophila. We found that Hand-Gal4>skd RNAi (KC) Drosophila had impaired cardiac function, metabolic impairment, and premature aging. Regular exercise significantly improved cardiac function and metabolism and delayed aging in HFD KC Drosophila. Thus, our study found that the effect of skd on cardiac energy metabolism in the heart affected cardiac function. Exercise may counteract age-related cardiac dysfunction and metabolic disturbances caused by HFD and heart-specific knockdown of skd. Skd may be a potential therapeutic target for heart disease.
Collapse
|
49
|
Simmons C, Bradshaw TW, Armstrong AR. Methods to Analyze Nutritional and Inter-Organ Control of Drosophila Ovarian Germline Stem Cells. Methods Mol Biol 2023; 2677:81-97. [PMID: 37464236 DOI: 10.1007/978-1-0716-3259-8_4] [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: 07/20/2023]
Abstract
Physiological status, particularly dietary input, has major impacts on the Drosophila melanogaster ovarian germline stem cell lineage. Moreover, several studies have shed light on the role that inter-organ communication plays in coordinating whole-organism responses to changes in physiology. For example, nutrient-sensing signaling pathways function within the fat body to regulate germline stem cells and their progeny in the ovary. Together with its incredible genetic and cell biological toolkits, Drosophila serves as an amenable model organism to use for uncovering molecular mechanisms that underlie physiological control of adult stem cells. In this methods chapter, we describe a general dietary manipulation paradigm, genetic manipulation of adult adipocytes, and whole-mount ovary immunofluorescence to investigate physiological control of germline stem cells.
Collapse
Affiliation(s)
- Chad Simmons
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
| | - Tancia W Bradshaw
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA
| | - Alissa R Armstrong
- Department of Biological Sciences, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
50
|
Cormier RJ, Doiron JA, Touaibia M, Surette ME, Pichaud N. Time-dependent metabolome and fatty acid profile changes following a high-fat diet exposure in Drosophila melanogaster. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 152:103892. [PMID: 36493963 DOI: 10.1016/j.ibmb.2022.103892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/29/2022] [Accepted: 12/05/2022] [Indexed: 06/17/2023]
Abstract
High-fat diets (HFDs) are often used to study metabolic disorders using different animal models. However, the underlying cellular mechanisms pertaining to the concurrent loss of metabolic homeostasis characteristics of these disorders are still unclear mainly because the effects of such diets are also dependent on the time frame of the experiments. Here, we used the fruit fly, Drosophila melanogaster, to investigate the metabolic dynamic effects following 0, 2, 4, 7 and 9 days of an exposure to a HFD (standard diet supplemented with 20% w/v coconut oil, rich in 12:0 and 14:0) by combining NMR metabolomics and GC-FID fatty acid profiling. Our results show that after 2 days, the ingested 12:0 and 14:0 fatty acids are used for both lipogenesis and fatty acid oxidation. After 4 days, metabolites from several different pathways are highly modulated in response to the HFD, and an accumulation of 12:0 is also observed, suggesting that the balance of lipid, amino acid and carbohydrate metabolism is profoundly perturbed at this specific time point. Following a longer exposure to the HFD (and notably after 9 days), an accumulation of many metabolites is observed indicating a clear dysfunction of the metabolic system. Overall, our study highlights the relevance of the Drosophila model to study metabolic disorders and the importance of the duration of the exposure to a HFD to study the dynamics of the fundamental mechanisms that control metabolism following exposure to dietary fats. This knowledge is crucial to understand the development and progression of metabolic diseases.
Collapse
Affiliation(s)
- Robert J Cormier
- New Brunswick Centre for Precision Medicine, Moncton, NB, E1A 3E9, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1 A 3E9, Canada
| | - Jeremie A Doiron
- New Brunswick Centre for Precision Medicine, Moncton, NB, E1A 3E9, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1 A 3E9, Canada
| | - Mohamed Touaibia
- Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1 A 3E9, Canada
| | - Marc E Surette
- New Brunswick Centre for Precision Medicine, Moncton, NB, E1A 3E9, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1 A 3E9, Canada
| | - Nicolas Pichaud
- New Brunswick Centre for Precision Medicine, Moncton, NB, E1A 3E9, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, NB, E1 A 3E9, Canada.
| |
Collapse
|