1
|
Pales Espinosa E, Farhat S, Allam B. In silico identification of neuropeptide genes encoded by the genome of Crassostrea virginica with a special emphasis on feeding-related genes. Comp Biochem Physiol A Mol Integr Physiol 2024; 301:111792. [PMID: 39694410 DOI: 10.1016/j.cbpa.2024.111792] [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: 09/19/2024] [Revised: 12/11/2024] [Accepted: 12/11/2024] [Indexed: 12/20/2024]
Abstract
Suspension-feeding bivalves, including the oyster Crassostrea virginica, use mucosal lectins to capture food particles. For instance, oysters can increase the transcription of these molecules to enhance food uptake. However, the regulatory processes influencing food uptake remain unclear although likely involve neuropeptides. Information on the neuropeptidome of C. virginica is limited, hindering the comprehension of its physiology, including energy homeostasis. This study explored the genome of C. virginica to identify neuropeptide precursors in silico and compared these with orthologs from other mollusks. A special focus was given to genes with potential implication in feeding processes. qPCR was used to determine the main organs of transcription of feeding-related genes. To further probe the function of target neuropeptides, visceral ganglia extracts and synthetic NPF were injected into oysters to evaluate their impact on genes associated with feeding and energy homeostasis. A total of eighty-five neuropeptides genes were identified in C. virginica genome. About 50 % of these are suggested to play a role in feeding processes. qPCR analyses showed that visceral ganglia and digestive system are the main organs for the synthesis of feeding-related neuropeptides. Further, results showed that the transcription of several neuropeptide genes in the visceral ganglia, including NPF and insulin-like peptide, increased after starvation. Finally, the injection of visceral ganglia extracts and synthetic NPF increased the transcription of a mucosal lectin and a glycogen synthase, known to be involved in food capture and glucose storage. Overall, this study identifies key genes regulating oyster physiology, enhancing the understanding of the control of basic physiological mechanisms in C. virginica.
Collapse
Affiliation(s)
| | - Sarah Farhat
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY 11794-5000, USA; Institut Systématique Evolution Biodiversité (ISYEB), Muséum National d'Histoire Naturelle, CNRS, Sorbonne Université, EPHE, Université des Antilles, 57 rue Cuvier, 75005 Paris, France
| | - Bassem Allam
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, NY 11794-5000, USA
| |
Collapse
|
2
|
Nie P, Hu L, Feng X, Xu H. Gut Microbiota Disorders and Metabolic Syndrome: Tales of a Crosstalk Process. Nutr Rev 2024:nuae157. [PMID: 39504479 DOI: 10.1093/nutrit/nuae157] [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] [Indexed: 11/08/2024] Open
Abstract
The microbiota in humans consists of trillions of microorganisms that are involved in the regulation of the gastrointestinal tract and immune and metabolic homeostasis. The gut microbiota (GM) has a prominent impact on the pathogenesis of metabolic syndrome (MetS). This process is reciprocal, constituting a crosstalk process between the GM and MetS. In this review, GM directly or indirectly inducing MetS via the host-microbial metabolic axis has been systematically reviewed. Additionally, the specifically altered GM in MetS are detailed in this review. Moreover, short-chain fatty acids (SCFAs), as unique gut microbial metabolites, have a remarkable effect on MetS, and the role of SCFAs in MetS-related diseases is highlighted to supplement the gaps in this area. Finally, the existing therapeutics are outlined, and the superiority and shortcomings of different therapeutic approaches are discussed, in hopes that this review can contribute to the development of potential treatment strategies.
Collapse
Affiliation(s)
- Penghui Nie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Liehai Hu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Xiaoyan Feng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
| | - Hengyi Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China
- International Institute of Food Innovation Co., Ltd, Nanchang University, Nanchang 330200, China
| |
Collapse
|
3
|
Marco HG, Glendinning S, Ventura T, Gäde G. The gonadotropin-releasing hormone (GnRH) superfamily across Pancrustacea/Tetraconata: A role in metabolism? Mol Cell Endocrinol 2024; 590:112238. [PMID: 38616035 DOI: 10.1016/j.mce.2024.112238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 04/05/2024] [Indexed: 04/16/2024]
Affiliation(s)
- Heather G Marco
- Department of Biological Sciences, University of Cape Town, Rondebosch, 7701, South Africa.
| | - Susan Glendinning
- Centre for BioInnovation, University of the Sunshine Coast, Sippy Downs, Queensland, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, 4556, Australia
| | - Tomer Ventura
- Centre for BioInnovation, University of the Sunshine Coast, Sippy Downs, Queensland, 4556, Australia; School of Science, Technology and Engineering, University of the Sunshine Coast, Sippy Downs, Queensland, 4556, Australia
| | - Gerd Gäde
- Department of Biological Sciences, University of Cape Town, Rondebosch, 7701, South Africa
| |
Collapse
|
4
|
Ott RK, Williams IH, Armstrong AR. Improved whole-mount immunofluorescence protocol for consistent and robust labeling of adult Drosophila melanogaster adipose tissue. Biol Open 2024; 13:bio060491. [PMID: 39041865 DOI: 10.1242/bio.060491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024] Open
Abstract
Energy storage and endocrine functions of the Drosophila fat body make it an excellent model for elucidating mechanisms that underlie physiological and pathophysiological organismal metabolism. Combined with Drosophila's robust genetic and immunofluorescence microscopy toolkits, studies of Drosophila fat body function are ripe for cell biological analysis. Unlike the larval fat body, which is easily removed as a single, cohesive sheet of tissue, isolating intact adult fat body proves to be more challenging, thus hindering consistent immunofluorescence labeling even within a single piece of adipose tissue. Here, we describe an improved approach to handling Drosophila abdomens that ensures full access of the adult fat body to solutions generally used in immunofluorescence labeling protocols. In addition, we assess the quality of fluorescence reporter expression and antibody immunoreactivity in response to variations in fixative type, fixation incubation time, and detergent used for cellular permeabilization. Overall, we provide several recommendations for steps in a whole-mount staining protocol that results in consistent and robust immunofluorescence labeling of the adult Drosophila fat body.
Collapse
Affiliation(s)
- Rachael K Ott
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29072, USA
| | - Isaiah H Williams
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29072, USA
| | - Alissa R Armstrong
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29072, USA
| |
Collapse
|
5
|
Kubrak O, Jørgensen AF, Koyama T, Lassen M, Nagy S, Hald J, Mazzoni G, Madsen D, Hansen JB, Larsen MR, Texada MJ, Hansen JL, Halberg KV, Rewitz K. LGR signaling mediates muscle-adipose tissue crosstalk and protects against diet-induced insulin resistance. Nat Commun 2024; 15:6126. [PMID: 39033139 PMCID: PMC11271308 DOI: 10.1038/s41467-024-50468-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: 09/04/2023] [Accepted: 07/04/2024] [Indexed: 07/23/2024] Open
Abstract
Obesity impairs tissue insulin sensitivity and signaling, promoting type-2 diabetes. Although improving insulin signaling is key to reversing diabetes, the multi-organ mechanisms regulating this process are poorly defined. Here, we screen the secretome and receptome in Drosophila to identify the hormonal crosstalk affecting diet-induced insulin resistance and obesity. We discover a complex interplay between muscle, neuronal, and adipose tissues, mediated by Bone Morphogenetic Protein (BMP) signaling and the hormone Bursicon, that enhances insulin signaling and sugar tolerance. Muscle-derived BMP signaling, induced by sugar, governs neuronal Bursicon signaling. Bursicon, through its receptor Rickets, a Leucine-rich-repeat-containing G-protein coupled receptor (LGR), improves insulin secretion and insulin sensitivity in adipose tissue, mitigating hyperglycemia. In mouse adipocytes, loss of the Rickets ortholog LGR4 blunts insulin responses, showing an essential role of LGR4 in adipocyte insulin sensitivity. Our findings reveal a muscle-neuronal-fat-tissue axis driving metabolic adaptation to high-sugar conditions, identifying LGR4 as a critical mediator in this regulatory network.
Collapse
Affiliation(s)
- Olga Kubrak
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Anne F Jørgensen
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
- Novo Nordisk, Novo Nordisk Park, 2760, Maaløv, Denmark
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Mette Lassen
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Stanislav Nagy
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Jacob Hald
- Novo Nordisk, Novo Nordisk Park, 2760, Maaløv, Denmark
| | | | - Dennis Madsen
- Novo Nordisk, Novo Nordisk Park, 2760, Maaløv, Denmark
| | - Jacob B Hansen
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, 5230, Odense, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | | | - Kenneth V Halberg
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100, Copenhagen O, Denmark.
| |
Collapse
|
6
|
Meschi E, Duquenoy L, Otto N, Dempsey G, Waddell S. Compensatory enhancement of input maintains aversive dopaminergic reinforcement in hungry Drosophila. Neuron 2024; 112:2315-2332.e8. [PMID: 38795709 DOI: 10.1016/j.neuron.2024.04.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 03/12/2024] [Accepted: 04/30/2024] [Indexed: 05/28/2024]
Abstract
Hungry animals need compensatory mechanisms to maintain flexible brain function, while modulation reconfigures circuits to prioritize resource seeking. In Drosophila, hunger inhibits aversively reinforcing dopaminergic neurons (DANs) to permit the expression of food-seeking memories. Multitasking the reinforcement system for motivation potentially undermines aversive learning. We find that chronic hunger mildly enhances aversive learning and that satiated-baseline and hunger-enhanced learning require endocrine adipokinetic hormone (AKH) signaling. Circulating AKH influences aversive learning via its receptor in four neurons in the ventral brain, two of which are octopaminergic. Connectomics revealed AKH receptor-expressing neurons to be upstream of several classes of ascending neurons, many of which are presynaptic to aversively reinforcing DANs. Octopaminergic modulation of and output from at least one of these ascending pathways is required for shock- and bitter-taste-reinforced aversive learning. We propose that coordinated enhancement of input compensates for hunger-directed inhibition of aversive DANs to preserve reinforcement when required.
Collapse
Affiliation(s)
- Eleonora Meschi
- University of Oxford, Centre for Neural Circuits and Behaviour, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Lucille Duquenoy
- University of Oxford, Centre for Neural Circuits and Behaviour, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Nils Otto
- University of Oxford, Centre for Neural Circuits and Behaviour, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Georgia Dempsey
- University of Oxford, Centre for Neural Circuits and Behaviour, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK
| | - Scott Waddell
- University of Oxford, Centre for Neural Circuits and Behaviour, Tinsley Building, Mansfield Road, Oxford OX1 3SR, UK.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Gao J, Zhang S, Deng P, Wu Z, Lemaitre B, Zhai Z, Guo Z. Dietary L-Glu sensing by enteroendocrine cells adjusts food intake via modulating gut PYY/NPF secretion. Nat Commun 2024; 15:3514. [PMID: 38664401 PMCID: PMC11045819 DOI: 10.1038/s41467-024-47465-4] [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: 02/09/2023] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Amino acid availability is monitored by animals to adapt to their nutritional environment. Beyond gustatory receptors and systemic amino acid sensors, enteroendocrine cells (EECs) are believed to directly percept dietary amino acids and secrete regulatory peptides. However, the cellular machinery underlying amino acid-sensing by EECs and how EEC-derived hormones modulate feeding behavior remain elusive. Here, by developing tools to specifically manipulate EECs, we find that Drosophila neuropeptide F (NPF) from mated female EECs inhibits feeding, similar to human PYY. Mechanistically, dietary L-Glutamate acts through the metabotropic glutamate receptor mGluR to decelerate calcium oscillations in EECs, thereby causing reduced NPF secretion via dense-core vesicles. Furthermore, two dopaminergic enteric neurons expressing NPFR perceive EEC-derived NPF and relay an anorexigenic signal to the brain. Thus, our findings provide mechanistic insights into how EECs assess food quality and identify a conserved mode of action that explains how gut NPF/PYY modulates food intake.
Collapse
Affiliation(s)
- Junjun Gao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Deng
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
- Department of Mechanical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Zhigang Wu
- State Key Laboratory of Digital Manufacturing Equipment and Technology, Huazhong University of Science and Technology, Wuhan, PR China
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Zongzhao Zhai
- State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, Hunan, PR China.
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Zandawala M, Gera J. Leptin- and cytokine-like unpaired signaling in Drosophila. Mol Cell Endocrinol 2024; 584:112165. [PMID: 38266772 DOI: 10.1016/j.mce.2024.112165] [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: 11/15/2023] [Revised: 01/13/2024] [Accepted: 01/19/2024] [Indexed: 01/26/2024]
Abstract
Animals have evolved a multitude of signaling pathways that enable them to orchestrate diverse physiological processes to tightly regulate systemic homeostasis. This signaling is mediated by various families of peptide hormones and cytokines that are conserved across the animal kingdom. In this review, we primarily focus on the unpaired (Upd) family of proteins in Drosophila which are evolutionarily related to mammalian leptin and the cytokine interleukin 6. We summarize expression patterns of Upd in Drosophila and discuss the parallels in structure, signaling pathway, and functions between Upd and their mammalian counterparts. In particular, we focus on the roles of Upd in governing metabolic homeostasis, growth and development, and immune responses. We aim to stimulate future studies on leptin-like signaling in other phyla which can help bridge the evolutionary gap between insect Upd and vertebrate leptin and cytokines like interleukin 6.
Collapse
Affiliation(s)
- Meet Zandawala
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany; Department of Biochemistry and Molecular Biology, University of Nevada, Reno, NV, 89557, USA.
| | - Jayati Gera
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, 97074, Würzburg, Germany
| |
Collapse
|
11
|
Zhang P, Catterson JH, Grönke S, Partridge L. Inhibition of S6K lowers age-related inflammation and increases lifespan through the endolysosomal system. NATURE AGING 2024; 4:491-509. [PMID: 38413780 PMCID: PMC11031405 DOI: 10.1038/s43587-024-00578-3] [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: 09/09/2022] [Accepted: 01/24/2024] [Indexed: 02/29/2024]
Abstract
Suppression of target of rapamycin complex 1 (TORC1) by rapamycin ameliorates aging in diverse species. S6 kinase (S6K) is an essential mediator, but the mechanisms involved are unclear. Here we show that activation of S6K specifically in Drosophila fat-body blocked extension of lifespan by rapamycin, induced accumulation of multilamellar lysosomes and blocked age-associated hyperactivation of the NF-κB-like immune deficiency (IMD) pathway, indicative of reduced inflammaging. Syntaxin 13 mediated the effects of TORC1-S6K signaling on lysosome morphology and inflammaging, suggesting they may be linked. Inflammaging depended on the IMD receptor regulatory isoform PGRP-LC, and repression of the IMD pathway from midlife extended lifespan. Age-related inflammaging was higher in females than in males and was not lowered in males by rapamycin treatment or lowered S6K. Rapamycin treatment also elevated Syntaxin 12/13 levels in mouse liver and prevented age-related increase in noncanonical NF-κB signaling, suggesting that the effect of TORC1 on inflammaging is conserved from flies to mammals.
Collapse
Affiliation(s)
- Pingze Zhang
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - James H Catterson
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, London, UK.
| |
Collapse
|
12
|
Zhang S, Wu S, Yao R, Wei X, Ohlstein B, Guo Z. Eclosion muscles secrete ecdysteroids to initiate asymmetric intestinal stem cell division in Drosophila. Dev Cell 2024; 59:125-140.e12. [PMID: 38096823 DOI: 10.1016/j.devcel.2023.11.016] [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: 06/29/2023] [Revised: 10/05/2023] [Accepted: 11/14/2023] [Indexed: 01/11/2024]
Abstract
During organ development, tissue stem cells first expand via symmetric divisions and then switch to asymmetric divisions to minimize the time to obtain a mature tissue. In the Drosophila midgut, intestinal stem cells switch their divisions from symmetric to asymmetric at midpupal development to produce enteroendocrine cells. However, the signals that initiate this switch are unknown. Here, we identify the signal as ecdysteroids. In the presence of ecdysone, EcR and Usp promote the expression of E93 to suppress Br expression, resulting in asymmetric divisions. Surprisingly, the primary source of pupal ecdysone is not from the prothoracic gland but from dorsal internal oblique muscles (DIOMs), a group of transient skeletal muscles that are required for eclosion. Genetic analysis shows that DIOMs secrete ecdysteroids during mTOR-mediated muscle remodeling. Our findings identify sequential endocrine and mechanical roles for skeletal muscle, which ensure the timely asymmetric divisions of intestinal stem cells.
Collapse
Affiliation(s)
- Song Zhang
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Song Wu
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ruining Yao
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xueying Wei
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Benjamin Ohlstein
- Children's Research Institute and Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zheng Guo
- Department of Medical Genetics, School of Basic Medicine, Institute for Brain Research, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Cell Architecture Research Center, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China.
| |
Collapse
|
13
|
Holcombe J, Weavers H. Functional-metabolic coupling in distinct renal cell types coordinates organ-wide physiology and delays premature ageing. Nat Commun 2023; 14:8405. [PMID: 38110414 PMCID: PMC10728150 DOI: 10.1038/s41467-023-44098-x] [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: 05/26/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023] Open
Abstract
Precise coupling between cellular physiology and metabolism is emerging as a vital relationship underpinning tissue health and longevity. Nevertheless, functional-metabolic coupling within heterogenous microenvironments in vivo remains poorly understood due to tissue complexity and metabolic plasticity. Here, we establish the Drosophila renal system as a paradigm for linking mechanistic analysis of metabolism, at single-cell resolution, to organ-wide physiology. Kidneys are amongst the most energetically-demanding organs, yet exactly how individual cell types fine-tune metabolism to meet their diverse, unique physiologies over the life-course remains unclear. Integrating live-imaging of metabolite and organelle dynamics with spatio-temporal genetic perturbation within intact functional tissue, we uncover distinct cellular metabolic signatures essential to support renal physiology and healthy ageing. Cell type-specific programming of glucose handling, PPP-mediated glutathione regeneration and FA β-oxidation via dynamic lipid-peroxisomal networks, downstream of differential ERR receptor activity, precisely match cellular energetic demands whilst limiting damage and premature senescence; however, their dramatic dysregulation may underlie age-related renal dysfunction.
Collapse
Affiliation(s)
- Jack Holcombe
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK
| | - Helen Weavers
- School of Biochemistry, Biomedical Sciences, University of Bristol, Bristol, BS8 1TD, UK.
| |
Collapse
|
14
|
Fuse N, Hashiba H, Ishibashi K, Suzuki T, Nguyen QD, Fujii K, Ikeda-Ohtsubo W, Kitazawa H, Tanimoto H, Kurata S. Neural control of redox response and microbiota-triggered inflammation in Drosophila gut. Front Immunol 2023; 14:1268611. [PMID: 37965334 PMCID: PMC10642236 DOI: 10.3389/fimmu.2023.1268611] [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: 07/28/2023] [Accepted: 10/02/2023] [Indexed: 11/16/2023] Open
Abstract
Background The neural system plays a critical role in controlling gut immunity, and the gut microbiota contributes to this process. However, the roles and mechanisms of gut-brain-microbiota interactions remain unclear. To address this issue, we employed Drosophila as a model organism. We have previously shown that NP3253 neurons, which are connected to the brain and gut, are essential for resistance to oral bacterial infections. Here, we aimed to investigate the role of NP3253 neurons in the regulation of gut immunity. Methods We performed RNA-seq analysis of the adult Drosophila gut after genetically inactivating the NP3253 neurons. Flies were reared under oral bacterial infection and normal feeding conditions. In addition, we prepared samples under germ-free conditions to evaluate the role of the microbiota in gut gene expression. We knocked down the genes regulated by NP3253 neurons and examined their susceptibility to oral bacterial infections. Results We found that immune-related gene expression was upregulated in NP3253 neuron-inactivated flies compared to the control. However, this upregulation was abolished in axenic flies, suggesting that the immune response was abnormally activated by the microbiota in NP3253 neuron-inactivated flies. In addition, redox-related gene expression was downregulated in NP3253 neuron-inactivated flies, and this downregulation was also observed in axenic flies. Certain redox-related genes were required for resistance to oral bacterial infections, suggesting that NP3253 neurons regulate the redox responses for gut immunity in a microbiota-independent manner. Conclusion These results show that NP3253 neurons regulate the appropriate gene expression patterns in the gut and contribute to maintain homeostasis during oral infections.
Collapse
Affiliation(s)
- Naoyuki Fuse
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Haruka Hashiba
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kentaro Ishibashi
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuro Suzuki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Quang-Dat Nguyen
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kiho Fujii
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | | | - Haruki Kitazawa
- Graduate School of Agricultural Science, Tohoku University, Sendai, Japan
- The Division for the Establishment of Frontier Sciences of the Organization for Advanced Studies, Tohoku University, Sendai, Japan
| | - Hiromu Tanimoto
- The Division for the Establishment of Frontier Sciences of the Organization for Advanced Studies, Tohoku University, Sendai, Japan
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Shoichiro Kurata
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
- The Division for the Establishment of Frontier Sciences of the Organization for Advanced Studies, Tohoku University, Sendai, Japan
| |
Collapse
|
15
|
Nagai H, Nagai LAE, Tasaki S, Nakato R, Umetsu D, Kuranaga E, Miura M, Nakajima Y. Nutrient-driven dedifferentiation of enteroendocrine cells promotes adaptive intestinal growth in Drosophila. Dev Cell 2023; 58:1764-1781.e10. [PMID: 37689060 DOI: 10.1016/j.devcel.2023.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 05/05/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Post-developmental organ resizing improves organismal fitness under constantly changing nutrient environments. Although stem cell abundance is a fundamental determinant of adaptive resizing, our understanding of its underlying mechanisms remains primarily limited to the regulation of stem cell division. Here, we demonstrate that nutrient fluctuation induces dedifferentiation in the Drosophila adult midgut to drive adaptive intestinal growth. From lineage tracing and single-cell RNA sequencing, we identify a subpopulation of enteroendocrine (EE) cells that convert into functional intestinal stem cells (ISCs) in response to dietary glucose and amino acids by activating the JAK-STAT pathway. Genetic ablation of EE-derived ISCs severely impairs ISC expansion and midgut growth despite the retention of resident ISCs, and in silico modeling further indicates that EE dedifferentiation enables an efficient increase in the midgut cell number while maintaining epithelial cell composition. Our findings identify a physiologically induced dedifferentiation that ensures ISC expansion during adaptive organ growth in concert with nutrient conditions.
Collapse
Affiliation(s)
- Hiroki Nagai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| | | | - Sohei Tasaki
- Graduate School of Science, Hokkaido University, Sapporo, Hokkaido 060-0810, Japan
| | - Ryuichiro Nakato
- Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Daiki Umetsu
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Science, Osaka University, Osaka 560-0043, Japan
| | - Erina Kuranaga
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan
| | - Masayuki Miura
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan
| | - Yuichiro Nakajima
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan; Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi 980-0845, Japan.
| |
Collapse
|
16
|
Li J, Lyu B, Bi J, Shan R, Stanley D, Feng Q, Song Q. Partner of neuropeptide bursicon homodimer pburs mediates a novel antimicrobial peptide Ten3LP via Dif/Dorsal2 in Tribolium castaneum. Int J Biol Macromol 2023; 247:125840. [PMID: 37454995 DOI: 10.1016/j.ijbiomac.2023.125840] [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: 04/14/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Bursicon is a cystine knot family neuropeptide, composed of two subunits, bursicon (burs) and partner of burs (pburs). The subunits can form heterodimers to regulate cuticle tanning and wing maturation and homodimers to signal different biological functions in innate immunity, midgut stem cell proliferation and energy homeostasis, and reproductive physiology in the model insects Drosophila melanogaster or Tribolium castaneum. Here, we report on the role of the pburs homodimer in signaling innate immunity in T. castaneum larvae. Through transcriptome analysis we identified a set of immune-related genes that respond to pburs RNAi. Treating larvae with recombinant-pburs protein led to up-regulation of antimicrobial peptide (AMP) genes in vivo and in vitro. The upregulation of most AMP genes was dependent on the NF-κB transcription factor Relish. Most importantly, we identified a novel AMP, Tenecin 3-like peptide (Ten3LP), regulated by pburs via NF-κB transcription factor Dorsal-related immunity factor (Dif)/Dorsal2, but not Relish. We conducted Ten3LP RNAi, synthesized recombinant Ten3LP protein for microbial inhibition assays and functionally characterized Ten3LP as an AMP specific for fungi and Gram-positive bacteria. We demonstrate that expression of Ten3LP is activated by pburs via the Toll pathway. These findings identify new molecular targets for development of potential antibiotics for treating microbial infections and perhaps for RNAi based pest management technology.
Collapse
Affiliation(s)
- Jingjing Li
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA.
| | - Bo Lyu
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA.
| | - Jingxiu Bi
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA; Institution of Quality Standard and Testing Technology for Agro-product, Shandong Academy of Agricultural Science, Jinan, Shandong 250100, China.
| | - Ruiqi Shan
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA.
| | - David Stanley
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA; Biological Control of Insect Research Laboratory, United States Department of Agriculture-Agricultural Research Station (USDA/ARS), Columbia, MO 65203, USA.
| | - Qili Feng
- Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology, School of Life Sciences, South China Normal University, Guangzhou 510631, China.
| | - Qisheng Song
- Division of Plant Science and Technology, University of Missouri, Columbia, MO 65211, USA.
| |
Collapse
|
17
|
Li Y, Zhou X, Cheng C, Ding G, Zhao P, Tan K, Chen L, Perrimon N, Veenstra JA, Zhang L, Song W. Gut AstA mediates sleep deprivation-induced energy wasting in Drosophila. Cell Discov 2023; 9:49. [PMID: 37221172 DOI: 10.1038/s41421-023-00541-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/13/2023] [Indexed: 05/25/2023] Open
Abstract
Severe sleep deprivation (SD) has been highly associated with systemic energy wasting, such as lipid loss and glycogen depletion. Despite immune dysregulation and neurotoxicity observed in SD animals, whether and how the gut-secreted hormones participate in SD-induced disruption of energy homeostasis remains largely unknown. Using Drosophila as a conserved model organism, we characterize that production of intestinal Allatostatin A (AstA), a major gut-peptide hormone, is robustly increased in adult flies bearing severe SD. Interestingly, the removal of AstA production in the gut using specific drivers significantly improves lipid loss and glycogen depletion in SD flies without affecting sleep homeostasis. We reveal the molecular mechanisms whereby gut AstA promotes the release of an adipokinetic hormone (Akh), an insulin counter-regulatory hormone functionally equivalent to mammalian glucagon, to mobilize systemic energy reserves by remotely targeting its receptor AstA-R2 in Akh-producing cells. Similar regulation of glucagon secretion and energy wasting by AstA/galanin is also observed in SD mice. Further, integrating single-cell RNA sequencing and genetic validation, we uncover that severe SD results in ROS accumulation in the gut to augment AstA production via TrpA1. Altogether, our results demonstrate the essential roles of the gut-peptide hormone AstA in mediating SD-associated energy wasting.
Collapse
Affiliation(s)
- Yingge Li
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiaoya Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Guangming Ding
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Peng Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Kai Tan
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Lixia Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Norbert Perrimon
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Jan A Veenstra
- INCIA, UMR 5287 CNRS, University of Bordeaux, Talence, France
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Okamoto N, Watanabe A. Interorgan communication through peripherally derived peptide hormones in Drosophila. Fly (Austin) 2022; 16:152-176. [PMID: 35499154 PMCID: PMC9067537 DOI: 10.1080/19336934.2022.2061834] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/21/2022] [Accepted: 03/29/2022] [Indexed: 02/06/2023] Open
Abstract
In multicellular organisms, endocrine factors such as hormones and cytokines regulate development and homoeostasis through communication between different organs. For understanding such interorgan communications through endocrine factors, the fruit fly Drosophila melanogaster serves as an excellent model system due to conservation of essential endocrine systems between flies and mammals and availability of powerful genetic tools. In Drosophila and other insects, functions of neuropeptides or peptide hormones from the central nervous system have been extensively studied. However, a series of recent studies conducted in Drosophila revealed that peptide hormones derived from peripheral tissues also play critical roles in regulating multiple biological processes, including growth, metabolism, reproduction, and behaviour. Here, we summarise recent advances in understanding target organs/tissues and functions of peripherally derived peptide hormones in Drosophila and describe how these hormones contribute to various biological events through interorgan communications.
Collapse
Affiliation(s)
- Naoki Okamoto
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Akira Watanabe
- Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
19
|
Gáliková M, Klepsatel P. Ion transport peptide regulates energy intake, expenditure, and metabolic homeostasis in Drosophila. Genetics 2022; 222:iyac150. [PMID: 36190340 PMCID: PMC9713441 DOI: 10.1093/genetics/iyac150] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/21/2022] [Indexed: 12/13/2022] Open
Abstract
In mammals, energy homeostasis is regulated by the antagonistic action of hormones insulin and glucagon. However, in contrast to the highly conserved insulin, glucagon is absent in most invertebrates. Although there are several endocrine regulators of energy expenditure and catabolism (such as the adipokinetic hormone), no single invertebrate hormone with all of the functions of glucagon has been described so far. Here, we used genetic gain- and loss-of-function experiments to show that the Drosophila gene Ion transport peptide (ITP) codes for a novel catabolic regulator that increases energy expenditure, lowers fat and glycogen reserves, and increases glucose and trehalose. Intriguingly, Ion transport peptide has additional functions reminiscent of glucagon, such as inhibition of feeding and transit of the meal throughout the digestive tract. Furthermore, Ion transport peptide interacts with the well-known signaling via the Adipokinetic hormone; Ion transport peptide promotes the pathway by stimulating Adipokinetic hormone secretion and transcription of the receptor AkhR. The genetic manipulations of Ion transport peptide on standard and Adipokinetic hormone-deficient backgrounds showed that the Adipokinetic hormone peptide mediates the hyperglycemic and hypertrehalosemic effects of Ion transport peptide, while the other metabolic functions of Ion transport peptide seem to be Adipokinetic hormone independent. In addition, Ion transport peptide is necessary for critical processes such as development, starvation-induced foraging, reproduction, and average lifespan. Altogether, our work describes a novel master regulator of fly physiology with functions closely resembling mammalian glucagon.
Collapse
Affiliation(s)
- Martina Gáliková
- Institute of Zoology, Slovak Academy of Sciences, 845 06 Bratislava, Slovakia
- Department of Zoology, Stockholm University, 106 91 Stockholm, Sweden
| | - Peter Klepsatel
- Institute of Zoology, Slovak Academy of Sciences, 845 06 Bratislava, Slovakia
- Institute of Molecular Physiology and Genetics, Centre of Biosciences, Slovak Academy of Sciences, 840 05 Bratislava, Slovakia
| |
Collapse
|
20
|
Malita A, Kubrak O, Koyama T, Ahrentløv N, Texada MJ, Nagy S, Halberg KV, Rewitz K. A gut-derived hormone suppresses sugar appetite and regulates food choice in Drosophila. Nat Metab 2022; 4:1532-1550. [PMID: 36344765 PMCID: PMC9684077 DOI: 10.1038/s42255-022-00672-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 09/30/2022] [Indexed: 11/09/2022]
Abstract
Animals must adapt their dietary choices to meet their nutritional needs. How these needs are detected and translated into nutrient-specific appetites that drive food-choice behaviours is poorly understood. Here we show that enteroendocrine cells of the adult female Drosophila midgut sense nutrients and in response release neuropeptide F (NPF), which is an ortholog of mammalian neuropeptide Y-family gut-brain hormones. Gut-derived NPF acts on glucagon-like adipokinetic hormone (AKH) signalling to induce sugar satiety and increase consumption of protein-rich food, and on adipose tissue to promote storage of ingested nutrients. Suppression of NPF-mediated gut signalling leads to overconsumption of dietary sugar while simultaneously decreasing intake of protein-rich yeast. Furthermore, gut-derived NPF has a female-specific function in promoting consumption of protein-containing food in mated females. Together, our findings suggest that gut NPF-to-AKH signalling modulates specific appetites and regulates food choice to ensure homeostatic consumption of nutrients, providing insight into the hormonal mechanisms that underlie nutrient-specific hungers.
Collapse
Affiliation(s)
- Alina Malita
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Olga Kubrak
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Nadja Ahrentløv
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stanislav Nagy
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth V Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
21
|
De Backer JF, Grunwald Kadow IC. A role for glia in cellular and systemic metabolism: insights from the fly. CURRENT OPINION IN INSECT SCIENCE 2022; 53:100947. [PMID: 35772690 DOI: 10.1016/j.cois.2022.100947] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 06/15/2023]
Abstract
Excitability and synaptic transmission make neurons high-energy consumers. However, neurons do not store carbohydrates or lipids. Instead, they need support cells to fuel their metabolic demands. This role is assumed by glia, both in vertebrates and invertebrates. Many questions remain regarding the coupling between neuronal activity and energy demand on the one hand, and nutrient supply by glia on the other hand. Here, we review recent advances showing that fly glia, similar to their role in vertebrates, fuel neurons in times of high energetic demand, such as during memory formation and long-term storage. Vertebrate glia also play a role in the modulation of neurons, their communication, and behavior, including food search and feeding. We discuss recent literature pointing to similar roles of fly glia in behavior and metabolism.
Collapse
Affiliation(s)
- Jean-François De Backer
- Technical University of Munich, School of Life Sciences, Liesel-Beckmann-Str. 4, 85354 Freising, Germany; University of Bonn, Faculty of Medicine, UKB, Institute of Physiology II, Nussallee 11, 53115 Bonn, Germany
| | - Ilona C Grunwald Kadow
- Technical University of Munich, School of Life Sciences, Liesel-Beckmann-Str. 4, 85354 Freising, Germany; University of Bonn, Faculty of Medicine, UKB, Institute of Physiology II, Nussallee 11, 53115 Bonn, Germany.
| |
Collapse
|
22
|
Hauser F, Koch TL, Grimmelikhuijzen CJP. Review: The evolution of peptidergic signaling in Cnidaria and Placozoa, including a comparison with Bilateria. Front Endocrinol (Lausanne) 2022; 13:973862. [PMID: 36213267 PMCID: PMC9545775 DOI: 10.3389/fendo.2022.973862] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Bilateria have bilateral symmetry and are subdivided into Deuterostomia (animals like vertebrates) and Protostomia (animals like insects and mollusks). Neuropeptides occur in both Proto- and Deuterostomia and they are frequently structurally related across these two lineages. For example, peptides belonging to the oxytocin/vasopressin family exist in both clades. The same is true for the G protein-coupled receptors (GPCRs) of these peptides. These observations suggest that these neuropeptides and their GPCRs were already present in the common ancestor of Proto- and Deuterostomia, which lived about 700 million years ago (MYA). Furthermore, neuropeptides and their GPCRs occur in two early-branching phyla that diverged before the emergence of Bilateria: Cnidaria (animals like corals and sea anemones), and Placozoa (small disk-like animals, feeding on algae). The sequences of these neuropeptides and their GPCRs, however, are not closely related to those from Bilateria. In addition, cnidarian neuropeptides and their receptors are not closely related to those from Placozoa. We propose that the divergence times between Cnidaria, Placozoa, and Bilateria might be too long for recognizing sequence identities. Leucine-rich repeats-containing GPCRs (LGRs) are a special class of GPCRs that are characterized by a long N-terminus containing 10-20 leucine-rich domains, which are used for ligand binding. Among the ligands for LGRs are dimeric glycoprotein hormones, and insulin-like peptides, such as relaxin. LGRs have been found not only in Proto- and Deuterostomia, but also in early emerging phyla, such as Cnidaria and Placozoa. Humans have eight LGRs. In our current review, we have revisited the annotations of LGRs from the sea anemone Nematostella vectensis and the placozoan Trichoplax adhaerens. We identified 13 sea anemone LGRs and no less than 46 LGRs from T. adhaerens. All eight human LGRs appear to have orthologues in sea anemones and placozoans. LGRs and their ligands, therefore, have a long evolutionary history, going back to the common ancestor of Cnidaria and Placozoa.
Collapse
Affiliation(s)
- Frank Hauser
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Thomas L. Koch
- Section for Cell and Neurobiology, Department of Biology, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | |
Collapse
|
23
|
Fedele G, Loh SHY, Celardo I, Leal NS, Lehmann S, Costa AC, Martins LM. Suppression of intestinal dysfunction in a Drosophila model of Parkinson's disease is neuroprotective. NATURE AGING 2022; 2:317-331. [PMID: 37117744 DOI: 10.1038/s43587-022-00194-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 02/16/2022] [Indexed: 04/30/2023]
Abstract
The innate immune response mounts a defense against foreign invaders and declines with age. An inappropriate induction of this response can cause diseases. Previous studies showed that mitochondria can be repurposed to promote inflammatory signaling. Damaged mitochondria can also trigger inflammation and promote diseases. Mutations in pink1, a gene required for mitochondrial health, cause Parkinson's disease, and Drosophila melanogaster pink1 mutants accumulate damaged mitochondria. Here, we show that defective mitochondria in pink1 mutants activate Relish targets and demonstrate that inflammatory signaling causes age-dependent intestinal dysfunction in pink1-mutant flies. These effects result in the death of intestinal cells, metabolic reprogramming and neurotoxicity. We found that Relish signaling is activated downstream of a pathway stimulated by cytosolic DNA. Suppression of Relish in the intestinal midgut of pink1-mutant flies restores mitochondrial function and is neuroprotective. We thus conclude that gut-brain communication modulates neurotoxicity in a fly model of Parkinson's disease through a mechanism involving mitochondrial dysfunction.
Collapse
Affiliation(s)
- Giorgio Fedele
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | | | - Ivana Celardo
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | | | - Susann Lehmann
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Ana C Costa
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
24
|
Medina A, Bellec K, Polcowñuk S, Cordero JB. Investigating local and systemic intestinal signalling in health and disease with Drosophila. Dis Model Mech 2022; 15:274860. [PMID: 35344037 PMCID: PMC8990086 DOI: 10.1242/dmm.049332] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Whole-body health relies on complex inter-organ signalling networks that enable organisms to adapt to environmental perturbations and to changes in tissue homeostasis. The intestine plays a major role as a signalling centre by producing local and systemic signals that are relayed to the body and that maintain intestinal and organismal homeostasis. Consequently, disruption of intestinal homeostasis and signalling are associated with systemic diseases and multi-organ dysfunction. In recent years, the fruit fly Drosophila melanogaster has emerged as a prime model organism to study tissue-intrinsic and systemic signalling networks of the adult intestine due to its genetic tractability and functional conservation with mammals. In this Review, we highlight Drosophila research that has contributed to our understanding of how the adult intestine interacts with its microenvironment and with distant organs. We discuss the implications of these findings for understanding intestinal and whole-body pathophysiology, and how future Drosophila studies might advance our knowledge of the complex interplay between the intestine and the rest of the body in health and disease. Summary: We outline work in the fruit fly Drosophila melanogaster that has contributed knowledge on local and whole-body signalling coordinated by the adult intestine, and discuss its implications in intestinal pathophysiology and associated systemic dysfunction.
Collapse
Affiliation(s)
- Andre Medina
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Karen Bellec
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Sofia Polcowñuk
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| | - Julia B Cordero
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK.,CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| |
Collapse
|
25
|
Nutrient Sensing via Gut in Drosophila melanogaster. Int J Mol Sci 2022; 23:ijms23052694. [PMID: 35269834 PMCID: PMC8910450 DOI: 10.3390/ijms23052694] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Nutrient-sensing mechanisms in animals' sense available nutrients to generate a physiological regulatory response involving absorption, digestion, and regulation of food intake and to maintain glucose and energy homeostasis. During nutrient sensing via the gastrointestinal tract, nutrients interact with receptors on the enteroendocrine cells in the gut, which in return respond by secreting various hormones. Sensing of nutrients by the gut plays a critical role in transmitting food-related signals to the brain and other tissues informing the composition of ingested food to digestive processes. These signals modulate feeding behaviors, food intake, metabolism, insulin secretion, and energy balance. The increasing significance of fly genetics with the availability of a vast toolbox for studying physiological function, expression of chemosensory receptors, and monitoring the gene expression in specific cells of the intestine makes the fly gut the most useful tissue for studying the nutrient-sensing mechanisms. In this review, we emphasize on the role of Drosophila gut in nutrient-sensing to maintain metabolic homeostasis and gut-brain cross talk using endocrine and neuronal signaling pathways stimulated by internal state or the consumption of various dietary nutrients. Overall, this review will be useful in understanding the post-ingestive nutrient-sensing mechanisms having a physiological and pathological impact on health and diseases.
Collapse
|
26
|
The gut hormone Allatostatin C/Somatostatin regulates food intake and metabolic homeostasis under nutrient stress. Nat Commun 2022; 13:692. [PMID: 35121731 PMCID: PMC8816919 DOI: 10.1038/s41467-022-28268-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
AbstractThe intestine is a central regulator of metabolic homeostasis. Dietary inputs are absorbed through the gut, which senses their nutritional value and relays hormonal information to other organs to coordinate systemic energy balance. However, the gut-derived hormones affecting metabolic and behavioral responses are poorly defined. Here we show that the endocrine cells of the Drosophila gut sense nutrient stress through a mechanism that involves the TOR pathway and in response secrete the peptide hormone allatostatin C, a Drosophila somatostatin homolog. Gut-derived allatostatin C induces secretion of glucagon-like adipokinetic hormone to coordinate food intake and energy mobilization. Loss of gut Allatostatin C or its receptor in the adipokinetic-hormone-producing cells impairs lipid and sugar mobilization during fasting, leading to hypoglycemia. Our findings illustrate a nutrient-responsive endocrine mechanism that maintains energy homeostasis under nutrient-stress conditions, a function that is essential to health and whose failure can lead to metabolic disorders.
Collapse
|
27
|
Lin HH, Kuang MC, Hossain I, Xuan Y, Beebe L, Shepherd AK, Rolandi M, Wang JW. A nutrient-specific gut hormone arbitrates between courtship and feeding. Nature 2022; 602:632-638. [PMID: 35140404 PMCID: PMC9271372 DOI: 10.1038/s41586-022-04408-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 12/22/2021] [Indexed: 11/08/2022]
Abstract
Animals must set behavioural priority in a context-dependent manner and switch from one behaviour to another at the appropriate moment1-3. Here we probe the molecular and neuronal mechanisms that orchestrate the transition from feeding to courtship in Drosophila melanogaster. We find that feeding is prioritized over courtship in starved males, and the consumption of protein-rich food rapidly reverses this order within a few minutes. At the molecular level, a gut-derived, nutrient-specific neuropeptide hormone-Diuretic hormone 31 (Dh31)-propels a switch from feeding to courtship. We further address the underlying kinetics with calcium imaging experiments. Amino acids from food acutely activate Dh31+ enteroendocrine cells in the gut, increasing Dh31 levels in the circulation. In addition, three-photon functional imaging of intact flies shows that optogenetic stimulation of Dh31+ enteroendocrine cells rapidly excites a subset of brain neurons that express Dh31 receptor (Dh31R). Gut-derived Dh31 excites the brain neurons through the circulatory system within a few minutes, in line with the speed of the feeding-courtship behavioural switch. At the circuit level, there are two distinct populations of Dh31R+ neurons in the brain, with one population inhibiting feeding through allatostatin-C and the other promoting courtship through corazonin. Together, our findings illustrate a mechanism by which the consumption of protein-rich food triggers the release of a gut hormone, which in turn prioritizes courtship over feeding through two parallel pathways.
Collapse
Affiliation(s)
- Hui-Hao Lin
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Meihua Christina Kuang
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Imran Hossain
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Yinan Xuan
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Laura Beebe
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Andrew K Shepherd
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Marco Rolandi
- Department of Electrical and Computer Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Jing W Wang
- Neurobiology Section, Division of Biological Sciences, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
28
|
Parra-Peralbo E, Talamillo A, Barrio R. Origin and Development of the Adipose Tissue, a Key Organ in Physiology and Disease. Front Cell Dev Biol 2022; 9:786129. [PMID: 34993199 PMCID: PMC8724577 DOI: 10.3389/fcell.2021.786129] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/01/2021] [Indexed: 12/17/2022] Open
Abstract
Adipose tissue is a dynamic organ, well known for its function in energy storage and mobilization according to nutrient availability and body needs, in charge of keeping the energetic balance of the organism. During the last decades, adipose tissue has emerged as the largest endocrine organ in the human body, being able to secrete hormones as well as inflammatory molecules and having an important impact in multiple processes such as adipogenesis, metabolism and chronic inflammation. However, the cellular progenitors, development, homeostasis and metabolism of the different types of adipose tissue are not fully known. During the last decade, Drosophila melanogaster has demonstrated to be an excellent model to tackle some of the open questions in the field of metabolism and development of endocrine/metabolic organs. Discoveries ranged from new hormones regulating obesity to subcellular mechanisms that regulate lipogenesis and lipolysis. Here, we review the available evidences on the development, types and functions of adipose tissue in Drosophila and identify some gaps for future research. This may help to understand the cellular and molecular mechanism underlying the pathophysiology of this fascinating key tissue, contributing to establish this organ as a therapeutic target.
Collapse
Affiliation(s)
| | - Ana Talamillo
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC BioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| |
Collapse
|
29
|
Hughson BN. PKG acts in the adult corpora cardiaca to regulate nutrient stress-responsivity through adipokinetic hormone. JOURNAL OF INSECT PHYSIOLOGY 2022; 136:104339. [PMID: 34856210 DOI: 10.1016/j.jinsphys.2021.104339] [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: 01/20/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 06/13/2023]
Abstract
In Drosophila melanogaster, the adipokinetic hormone (AKH) is a glucagon-like peptide that acts antagonistically with insulin-like peptides to maintain metabolic homeostasis. AKH is biosynthesized in and secreted from the corpora cardiaca (CC). This report describes a CC-specific role for dg2 - which encodes a cGMP-dependent protein kinase (PKG) - as a regulator of AKH during adulthood. Transcriptional silencing of dg2 during adulthood decreased starvation resistance, increased sucrose responsiveness, and decreased whole body lipid content. PKG protein was localized to CC cell membranes, and starvation caused a significant decrease in CC intracellular AKH content. Strikingly, reduced CC-dg2 expression caused a significant decrease in intracellular AKH content in adults fed ad libitum. This work demonstrated that dysregulation of CC-specific dg2 expression during adult life impaired metabolic homeostasis, and that dg2 acted in the CC to regulate systemic AKH activity.
Collapse
Affiliation(s)
- Bryon N Hughson
- Department of Ecology and Evolutionary Biology, University of Toronto, Ontario, Canada; University of Toronto, 27 King's College Circle, Toronto, Ontario M5S 3B2, Canada.
| |
Collapse
|
30
|
Hughson BN. The Glucagon-Like Adipokinetic Hormone in Drosophila melanogaster - Biosynthesis and Secretion. Front Physiol 2021; 12:710652. [PMID: 35002748 PMCID: PMC8733639 DOI: 10.3389/fphys.2021.710652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 09/16/2021] [Indexed: 11/13/2022] Open
Abstract
Metabolic homeostasis requires the precise regulation of circulating sugar titers. In mammals, homeostatic control of circulating sugar titers requires the coordinated secretion and systemic activities of glucagon and insulin. Metabolic homeostasis is similarly regulated in Drosophila melanogaster through the glucagon-like adipokinetic hormone (AKH) and the Drosophila insulin-like peptides (DILPs). In flies and mammals, glucagon and AKH are biosynthesized in and secreted from specialized endocrine cells. KATP channels borne on these cells respond to fluctuations in circulating glucose titers and thereby regulate glucagon secretion. The influence of glucagon in the pathogenesis of type 2 diabetes mellitus is now recognized, and a crucial mechanism that regulates glucagon secretion was reported nearly a decade ago. Ongoing efforts to develop D. melanogaster models for metabolic syndrome must build upon this seminal work. These efforts make a critical review of AKH physiology timely. This review focuses on AKH biosynthesis and the regulation of glucose-responsive AKH secretion through changes in CC cell electrical activity. Future directions for AKH research in flies are discussed, including the development of models for hyperglucagonemia and epigenetic inheritance of acquired metabolic traits. Many avenues of AKH physiology remain to be explored and thus present great potential for improving the utility of D. melanogaster in metabolic research.
Collapse
Affiliation(s)
- Bryon N. Hughson
- Department of Ecology and Evolutionary Biology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Li J, Zhu Z, Bi J, Feng Q, Beerntsen BT, Song Q. Neuropeptide Bursicon Influences Reproductive Physiology in Tribolium Castaneum. Front Physiol 2021; 12:717437. [PMID: 34744761 PMCID: PMC8567023 DOI: 10.3389/fphys.2021.717437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/20/2021] [Indexed: 11/13/2022] Open
Abstract
Bursicon is a neuropeptide belonging to the cystine knot family and is composed of burs and partner of burs (pburs) subunits. It can form heterodimers or homodimers to execute different biological functions. Bursicon heterodimers regulate cuticle sclerotization and wing maturation, whereas bursicon homodimers mediate innate immunity and midgut stem cell proliferation. A recent study has shown that bursicon potentially induces the expression of vitellogenin (Vg) in the black tiger shrimp Penaeus monodon; however, the underlying mechanism remains unknown. In this study, we investigated the role of bursicon in the reproductive physiology of the red flour beetle, Tribolium castaneum. The knockdown of burs, pburs, or its receptor T. castaneum rickets (Tcrk) in 2-day pupae significantly downregulated the expression levels of Vg1, Vg2, and Vg receptor (VgR) genes in females 3- and 5-day post-adult emergence, leading to abnormal oocytes with limited Vg content. The silencing of burs repressed the number of eggs laid and completely inhibited egg hatch, whereas the silencing of pburs dramatically decreased the number of eggs laid, hatch rate, and offspring larval size, and this RNA interference (RNAi) effects persisted to the next generation. Furthermore, the knockdown of burs or pburs downregulated the expression of the insulin/insulin-like signaling/target of rapamycin (TOR) signaling genes encoding insulin receptor (InR), protein kinase B (Akt), TOR, and ribosomal protein S6 kinase (S6K). Most importantly, the injection of recombinant pburs (r-pburs) protein was able to upregulate the expression of Vg, VgR, InR, Akt, TOR, S6K, JH synthesis (JHAMT), Methoprene-tolerant (Met), and Taiman (Tai) in normal females and rescue the expression of Vg and VgR in pburs RNAi females but failed to rescue Vg and VgR in Tcrk knockdown females. We infer that bursicon homodimers influence Vg expression via the receptor Tcrk, possibly by mediating the expression of the juvenile hormone (JH) and IIS/TOR pathway genes, thereby regulating reproduction in T. castaneum.
Collapse
Affiliation(s)
- Jingjing Li
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| | - Zidan Zhu
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States.,Guangdong Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou, China
| | - Jingxiu Bi
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States.,Institution of Quality Standard and Testing Technology for Agro-Product, Shandong Academy of Agricultural Science, Jinan, China
| | - Qili Feng
- Guangdong Key Laboratory of Insect Developmental Biology and Applied Technology, Guangzhou Key Laboratory of Insect Development Regulation and Application Research, Institute of Insect Science and Technology and School of Life Sciences, South China Normal University, Guangzhou, China
| | - Brenda T Beerntsen
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States.,Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, United States
| | - Qisheng Song
- Division of Plant Science and Technology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
32
|
Wat LW, Chowdhury ZS, Millington JW, Biswas P, Rideout EJ. Sex determination gene transformer regulates the male-female difference in Drosophila fat storage via the adipokinetic hormone pathway. eLife 2021; 10:e72350. [PMID: 34672260 PMCID: PMC8594944 DOI: 10.7554/elife.72350] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 10/07/2021] [Indexed: 12/17/2022] Open
Abstract
Sex differences in whole-body fat storage exist in many species. For example, Drosophila females store more fat than males. Yet, the mechanisms underlying this sex difference in fat storage remain incompletely understood. Here, we identify a key role for sex determination gene transformer (tra) in regulating the male-female difference in fat storage. Normally, a functional Tra protein is present only in females, where it promotes female sexual development. We show that loss of Tra in females reduced whole-body fat storage, whereas gain of Tra in males augmented fat storage. Tra's role in promoting fat storage was largely due to its function in neurons, specifically the Adipokinetic hormone (Akh)-producing cells (APCs). Our analysis of Akh pathway regulation revealed a male bias in APC activity and Akh pathway function, where this sex-biased regulation influenced the sex difference in fat storage by limiting triglyceride accumulation in males. Importantly, Tra loss in females increased Akh pathway activity, and genetically manipulating the Akh pathway rescued Tra-dependent effects on fat storage. This identifies sex-specific regulation of Akh as one mechanism underlying the male-female difference in whole-body triglyceride levels, and provides important insight into the conserved mechanisms underlying sexual dimorphism in whole-body fat storage.
Collapse
Affiliation(s)
- Lianna W Wat
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Zahid S Chowdhury
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Jason W Millington
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Puja Biswas
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, The University of British ColumbiaVancouverCanada
| |
Collapse
|
33
|
Yoshinari Y, Kosakamoto H, Kamiyama T, Hoshino R, Matsuoka R, Kondo S, Tanimoto H, Nakamura A, Obata F, Niwa R. The sugar-responsive enteroendocrine neuropeptide F regulates lipid metabolism through glucagon-like and insulin-like hormones in Drosophila melanogaster. Nat Commun 2021; 12:4818. [PMID: 34376687 PMCID: PMC8355161 DOI: 10.1038/s41467-021-25146-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 07/24/2021] [Indexed: 02/08/2023] Open
Abstract
The enteroendocrine cell (EEC)-derived incretins play a pivotal role in regulating the secretion of glucagon and insulins in mammals. Although glucagon-like and insulin-like hormones have been found across animal phyla, incretin-like EEC-derived hormones have not yet been characterised in invertebrates. Here, we show that the midgut-derived hormone, neuropeptide F (NPF), acts as the sugar-responsive, incretin-like hormone in the fruit fly, Drosophila melanogaster. Secreted NPF is received by NPF receptor in the corpora cardiaca and in insulin-producing cells. NPF-NPFR signalling resulted in the suppression of the glucagon-like hormone production and the enhancement of the insulin-like peptide secretion, eventually promoting lipid anabolism. Similar to the loss of incretin function in mammals, loss of midgut NPF led to significant metabolic dysfunction, accompanied by lipodystrophy, hyperphagia, and hypoglycaemia. These results suggest that enteroendocrine hormones regulate sugar-dependent metabolism through glucagon-like and insulin-like hormones not only in mammals but also in insects.
Collapse
Affiliation(s)
- Yuto Yoshinari
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Hina Kosakamoto
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
| | - Takumi Kamiyama
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Ryo Hoshino
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Rena Matsuoka
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shu Kondo
- Genetic Strains Research Center, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Hiromu Tanimoto
- Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Akira Nakamura
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
- Laboratory of Germline Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Fumiaki Obata
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
- Department of Genetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Laboratory for Nutritional Biology, RIKEN Center for Biosystems Dynamics Research, Kobe, Hyogo, Japan
- Laboratory of Molecular Cell Biology and Development, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- AMED-PRIME, Japan Agency for Medical Research and Development Chiyoda-ku, Tokyo, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan.
- AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan.
| |
Collapse
|
34
|
The Mechanism of Oral Melatonin Ameliorates Intestinal and Adipose Lipid Dysmetabolism Through Reducing Escherichia Coli-Derived Lipopolysaccharide. Cell Mol Gastroenterol Hepatol 2021; 12:1643-1667. [PMID: 34242820 PMCID: PMC8536535 DOI: 10.1016/j.jcmgh.2021.06.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Gut microbiota have been reported to be sensitive to circadian rhythms and host lipometabolism, respectively. Although melatonin-mediated beneficial efforts on many physiological sites have been revealed, the regulatory actions of oral melatonin on the communication between gut microbiota and host are still not clear. Angiopoietin-like 4 (ANGPTL4) has been shown to be strongly responsible for the regulation of systemic lipid metabolism. Herein, we identified that oral melatonin improved lipid dysmetabolism in ileum and epididymal white adipose tissue (eWAT) via gut microbiota and ileac ANGPTL4. METHODS Analyses of jet-lag (JL) mice, JL mice with oral melatonin administration (JL+MT), and the control for mRNA and protein expression regarding lipid uptake and accumulation in ileum and eWAT were made. Gut microbiome sequencing and experimental validation of target strains were included. Functional analysis of key factors/pathways in the various rodent models, including the depletion of gut microbiota, mono-colonization of Escherichia coli, and other genetic intervention was made. Analyses of transcriptional regulation and effects of melatonin on E coli-derived lipopolysaccharide (LPS) in vitro were made. RESULTS JL mice have a higher level of ileal lipid uptake, fat accumulation in eWAT, and lower level of circulating ANGPTL4 in comparison with the control mice. JL mice also showed a significantly higher abundance of E coli and LPS than the control mice. Conversely, oral melatonin supplementation remarkably reversed these phenotypes. The test of depletion of gut microbiota further demonstrated that oral melatonin-mediated improvements on lipometabolism in JL mice were dependent on the presence of gut microbiota. By mono-colonization of E coli, LPS has been determined to trigger these changes similar to JL. Furthermore, we found that LPS served as a pivotal link that contributed to activating toll-like receptor 4 (TLR4)/signal transducer and activator of transcription 3 (STAT3_/REV-ERBα) signaling to up-regulate nuclear factor interleukin-3-regulated protein (NFIL3) expression, resulting in increased lipid uptake in ileum. In MODE-K cells, the activation of NFIL3 has further been shown to inhibit ANGPTL4 transcription, which is closely associated with lipid uptake and transport in peripheral tissues. Finally, we confirmed that melatonin inhibited LPS via repressing the expression of LpxC in E coli. CONCLUSIONS Overall, oral melatonin decreased the quantity of E coli-generated LPS, which alleviated NFIL3-induced transcriptional inhibition of ANGPTL4 through TLR4/IL-22/STAT3 signaling in ileum, thereby resulting in the amelioration of ileal lipid intake and lower fat accumulation in eWAT. These results address a novel regulation of oral melatonin originating from gut microbiota to host distal tissues, suggesting that microbe-generated metabolites are potential therapies for melatonin-mediated improvement of circadian rhythm disruption and related metabolic syndrome.
Collapse
|
35
|
Kim SK, Tsao DD, Suh GSB, Miguel-Aliaga I. Discovering signaling mechanisms governing metabolism and metabolic diseases with Drosophila. Cell Metab 2021; 33:1279-1292. [PMID: 34139200 PMCID: PMC8612010 DOI: 10.1016/j.cmet.2021.05.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/30/2021] [Accepted: 05/25/2021] [Indexed: 12/18/2022]
Abstract
There has been rapid growth in the use of Drosophila and other invertebrate systems to dissect mechanisms governing metabolism. New assays and approaches to physiology have aligned with superlative genetic tools in fruit flies to provide a powerful platform for posing new questions, or dissecting classical problems in metabolism and disease genetics. In multiple examples, these discoveries exploit experimental advantages as-yet unavailable in mammalian systems. Here, we illustrate how fly studies have addressed long-standing questions in three broad areas-inter-organ signaling through hormonal or neural mechanisms governing metabolism, intestinal interoception and feeding, and the cellular and signaling basis of sexually dimorphic metabolism and physiology-and how these findings relate to human (patho)physiology. The imaginative application of integrative physiology and related approaches in flies to questions in metabolism is expanding, and will be an engine of discovery, revealing paradigmatic features of metabolism underlying human diseases and physiological equipoise in health.
Collapse
Affiliation(s)
- Seung K Kim
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine (Endocrinology), Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Deborah D Tsao
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Greg S B Suh
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 34141, South Korea.
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
36
|
Boumard B, Bardin AJ. An amuse-bouche of stem cell regulation: Underlying principles and mechanisms from adult Drosophila intestinal stem cells. Curr Opin Cell Biol 2021; 73:58-68. [PMID: 34217969 DOI: 10.1016/j.ceb.2021.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/27/2022]
Abstract
Stem cells have essential functions in the development and maintenance of our organs. Improper regulation of adult stem cells and tissue homeostasis can result in cancers and age-dependent decline. Therefore, understanding how tissue-specific stem cells can accurately renew tissues is an important aim of regenerative medicine. The Drosophila midgut harbors multipotent adult stem cells that are essential to renew the gut in homeostatic conditions and upon stress-induced regeneration. It is now a widely used model system to decipher regulatory mechanisms of stem cell biology. Here, we review recent findings on how adult intestinal stem cells differentiate, interact with their environment, and change during aging.
Collapse
Affiliation(s)
- Benjamin Boumard
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France
| | - Allison J Bardin
- Institut Curie, PSL Research University, CNRS UMR 3215, INSERM U934, Stem Cells and Tissue Homeostasis Group, Paris, France.
| |
Collapse
|
37
|
Guo X, Lv J, Xi R. The specification and function of enteroendocrine cells in Drosophila and mammals: a comparative review. FEBS J 2021; 289:4773-4796. [PMID: 34115929 DOI: 10.1111/febs.16067] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/26/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
Enteroendocrine cells (EECs) in both invertebrates and vertebrates derive from intestinal stem cells (ISCs) and are scattered along the digestive tract, where they function in sensing various environmental stimuli and subsequently secrete neurotransmitters or neuropeptides to regulate diverse biological and physiological processes. To fulfill these functions, EECs are specified into multiple subtypes that occupy specific gut regions. With advances in single-cell technology, organoid culture experimental systems, and CRISPR/Cas9-mediated genomic editing, rapid progress has been made toward characterization of EEC subtypes in mammals. Additionally, studies of genetic model organisms-especially Drosophila melanogaster-have also provided insights about the molecular processes underlying EEC specification from ISCs and about the establishment of diverse EEC subtypes. In this review, we compare the regulation of EEC specification and function in mammals and Drosophila, with a focus on EEC subtype characterization, on how internal and external regulators mediate EEC subtype specification, and on how EEC-mediated intra- and interorgan communications affect gastrointestinal physiology and pathology.
Collapse
Affiliation(s)
- Xingting Guo
- National Institute of Biological Sciences, Beijing, China
| | - Jiaying Lv
- National Institute of Biological Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Rongwen Xi
- National Institute of Biological Sciences, Beijing, China.,Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing, China
| |
Collapse
|
38
|
Le TD, Inoue YH. Sesamin Activates Nrf2/Cnc-Dependent Transcription in the Absence of Oxidative Stress in Drosophila Adult Brains. Antioxidants (Basel) 2021; 10:antiox10060924. [PMID: 34200419 PMCID: PMC8227698 DOI: 10.3390/antiox10060924] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 05/31/2021] [Accepted: 06/04/2021] [Indexed: 11/11/2022] Open
Abstract
Sesamin, a major lignin in sesame seeds, possesses health-promoting properties. Sesamin feeding suppresses several aging-related phenotypes such as age-dependent accumulation of damaged proteins in the muscles and neuronal loss in the brains of Drosophila adults with high levels of reactive oxygen species. Sesamin promotes the transcription of several genes that are responsible for oxidative stress, although the underlying mechanism remains unclear. Here, we aimed to demonstrate that sesamin mediates its action through activation of a transcription factor, Nrf2 (Cnc in Drosophila), essential for anti-aging oxidative stress response. Nrf2/Cnc activation was determined using the antioxidant response element, Green Fluorescence Protein reporter, that can monitor Nrf2/Cnc-dependent transcription. We observed strong fluorescence in the entire bodies, particularly in the abdomens and brains, of adult flies fed sesamin. Interestingly, Nrf2/Cnc was strongly activated in neuronal cells, especially in several neuron types, including glutamatergic and cholinergic, and some dopaminergic and/or serotonergic neurons but not in GABAergic neurons or the mushroom bodies of flies fed sesamin. These results indicate that the anti-aging effects of sesamin are exerted via activation of Nrf2/Cnc-dependent transcription to circumvent oxidative stress accumulation in several types of neurons of adult brains. Sesamin could be explored as a potential dietary supplement for preventing neurodegeneration associated with accumulation of oxidative stress.
Collapse
|
39
|
Heier C, Klishch S, Stilbytska O, Semaniuk U, Lushchak O. The Drosophila model to interrogate triacylglycerol biology. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158924. [PMID: 33716135 DOI: 10.1016/j.bbalip.2021.158924] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/24/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022]
Abstract
The deposition of storage fat in the form of triacylglycerol (TAG) is an evolutionarily conserved strategy to cope with fluctuations in energy availability and metabolic stress. Organismal TAG storage in specialized adipose tissues provides animals a metabolic reserve that sustains survival during development and starvation. On the other hand, excessive accumulation of adipose TAG, defined as obesity, is associated with an increasing prevalence of human metabolic diseases. During the past decade, the fruit fly Drosophila melanogaster, traditionally used in genetics and developmental biology, has been established as a versatile model system to study TAG metabolism and the etiology of lipid-associated metabolic diseases. Similar to humans, Drosophila TAG homeostasis relies on the interplay of organ systems specialized in lipid uptake, synthesis, and processing, which are integrated by an endocrine network of hormones and messenger molecules. Enzymatic formation of TAG from sugar or dietary lipid, its storage in lipid droplets, and its mobilization by lipolysis occur via mechanisms largely conserved between Drosophila and humans. Notably, dysfunctional Drosophila TAG homeostasis occurs in the context of aging, overnutrition, or defective gene function, and entails tissue-specific and organismal pathologies that resemble human disease. In this review, we summarize the physiology and biochemistry of TAG in Drosophila and outline the potential of this organism as a model system to understand the genetic and dietary basis of TAG storage and TAG-related metabolic disorders.
Collapse
Affiliation(s)
- Christoph Heier
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Humboldtstrasse 50, A-8010 Graz, Austria; BioTechMed-Graz, Graz, Austria.
| | - Svitlana Klishch
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Olha Stilbytska
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Uliana Semaniuk
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
40
|
Chatterjee N, Perrimon N. What fuels the fly: Energy metabolism in Drosophila and its application to the study of obesity and diabetes. SCIENCE ADVANCES 2021; 7:7/24/eabg4336. [PMID: 34108216 PMCID: PMC8189582 DOI: 10.1126/sciadv.abg4336] [Citation(s) in RCA: 87] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 04/23/2021] [Indexed: 05/16/2023]
Abstract
The organs and metabolic pathways involved in energy metabolism, and the process of ATP production from nutrients, are comparable between humans and Drosophila melanogaster This level of conservation, together with the power of Drosophila genetics, makes the fly a very useful model system to study energy homeostasis. Here, we discuss the major organs involved in energy metabolism in Drosophila and how they metabolize different dietary nutrients to generate adenosine triphosphate. Energy metabolism in these organs is controlled by cell-intrinsic, paracrine, and endocrine signals that are similar between Drosophila and mammals. We describe how these signaling pathways are regulated by several physiological and environmental cues to accommodate tissue-, age-, and environment-specific differences in energy demand. Last, we discuss several genetic and diet-induced fly models of obesity and diabetes that can be leveraged to better understand the molecular basis of these metabolic diseases and thereby promote the development of novel therapies.
Collapse
Affiliation(s)
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA.
- Howard Hughes Medical Institute, Boston, MA 02115, USA
| |
Collapse
|
41
|
Ben-Menahem D. GnRH-Related Neurohormones in the Fruit Fly Drosophila melanogaster. Int J Mol Sci 2021; 22:ijms22095035. [PMID: 34068603 PMCID: PMC8126107 DOI: 10.3390/ijms22095035] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 04/30/2021] [Accepted: 05/06/2021] [Indexed: 11/16/2022] Open
Abstract
Genomic and phylogenetic analyses of various invertebrate phyla revealed the existence of genes that are evolutionarily related to the vertebrate’s decapeptide gonadotropin-releasing hormone (GnRH) and the GnRH receptor genes. Upon the characterization of these gene products, encoding peptides and putative receptors, GnRH-related peptides and their G-protein coupled receptors have been identified. These include the adipokinetic hormone (AKH) and corazonin (CRZ) in insects and their cognate receptors that pair to form bioactive signaling systems, which network with additional neurotransmitters/hormones (e.g., octopamine and ecdysone). Multiple studies in the past 30 years have identified many aspects of the biology of these peptides that are similar in size to GnRH and function as neurohormones. This review briefly describes the main activities of these two neurohormones and their receptors in the fruit fly Drosophila melanogaster. The similarities and differences between Drosophila AKH/CRZ and mammalian GnRH signaling systems are discussed. Of note, while GnRH has a key role in reproduction, AKH and CRZ show pleiotropic activities in the adult fly, primarily in metabolism and stress responses. From a protein evolution standpoint, the GnRH/AKH/CRZ family nicely demonstrates the developmental process of neuropeptide signaling systems emerging from a putative common ancestor and leading to divergent activities in distal phyla.
Collapse
Affiliation(s)
- David Ben-Menahem
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
42
|
Singh A, Agrawal N. Deciphering the key mechanisms leading to alteration of lipid metabolism in Drosophila model of Huntington's disease. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166127. [PMID: 33722743 DOI: 10.1016/j.bbadis.2021.166127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/27/2021] [Accepted: 03/03/2021] [Indexed: 12/19/2022]
Abstract
Huntington's disease (HD) is an inherited, progressively debilitating disorder marked by prominent degeneration in striatal and cortical brain regions. HD is caused by (CAG)n repeat expansion in huntingtin (HTT) gene that translates into a mutant form of the ubiquitously present Huntingtin (HTT) protein. Extensive metabolic dysfunction coexisting with overt neuropathies has been evidenced in clinical and experimental settings of HD. Body weight loss despite normal to high caloric intake remains a critical determinant of the disease progression and a challenge for therapeutic interventions. In the present study, we intended to monitor the cellular and molecular perturbations in Drosophila, caused by pan-neuronal expression of mHTT (mutant Huntingtin) protein. We found aberrant transcription profile of key lipolytic and lipogenic genes in whole-body of the fly with disease progression. Interestingly, fatbody undergoes extensive alteration of vital cellular processes and eventually surrenders to increased apoptotic cell death in terminal stage of the disease. Extensive mitochondrial dysfunction from early disease stage along with calcium derangement at terminal stage were observed in fatbody, which contribute to its deteriorating integrity. All the mechanisms were monitored progressively, at different disease stages, and many alterations were documented in the early stage itself. Our study hence provides insight into the mechanisms through which neuronal expression of mHTT might be inflicting the profound systemic effects, specifically on lipid metabolism, and may open new therapeutic avenues for alleviation of the multidimensional disease.
Collapse
Affiliation(s)
- Akanksha Singh
- Department of Zoology, University of Delhi, Delhi 110007, India
| | - Namita Agrawal
- Department of Zoology, University of Delhi, Delhi 110007, India.
| |
Collapse
|
43
|
Wang Y, Wu X, Wang Z, Chen T, Zhou S, Chen J, Pang L, Ye X, Shi M, Huang J, Chen X. Symbiotic bracovirus of a parasite manipulates host lipid metabolism via tachykinin signaling. PLoS Pathog 2021; 17:e1009365. [PMID: 33647060 PMCID: PMC7951984 DOI: 10.1371/journal.ppat.1009365] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 03/11/2021] [Accepted: 02/09/2021] [Indexed: 12/31/2022] Open
Abstract
Parasites alter host energy homeostasis for their own development, but the mechanisms underlying this phenomenon remain largely unknown. Here, we show that Cotesia vestalis, an endoparasitic wasp of Plutella xylostella larvae, stimulates a reduction of host lipid levels. This process requires excess secretion of P. xylostella tachykinin (PxTK) peptides from enteroendocrine cells (EEs) in the midgut of the parasitized host larvae. We found that parasitization upregulates PxTK signaling to suppress lipogenesis in midgut enterocytes (ECs) in a non-cell-autonomous manner, and the reduced host lipid level benefits the development of wasp offspring and their subsequent parasitic ability. We further found that a C. vestalis bracovirus (CvBV) gene, CvBV 9–2, is responsible for PxTK induction, which in turn reduces the systemic lipid level of the host. Taken together, these findings illustrate a novel mechanism for parasite manipulation of host energy homeostasis by a symbiotic bracovirus gene to promote the development and increase the parasitic efficiency of an agriculturally important wasp species. Parasitic wasps are ubiquitous on earth and diverse. They lay eggs in or on the bodies of their hosts, and they have evolved adaptive strategies to regulate the energy metabolism of their hosts to match their own specific nutrition requirements. Here, we found that Cotesia vestalis, a solitary endoparasitoid of Plutella xylostella, uses symbiotic bracovirus as a weapon to manipulate host systemic lipid levels. Specifically, a C. vestalis bracovirus (CvBV) gene, CvBV 9–2, is responsible for the induction of PxTK, which in turn suppresses lipogenesis in the midgut of the parasitized host, leading to a nutritional lipid level suitable for the development and subsequent parasitic efficiency of C. vestalis wasps. Our study provides innovative insights into the mechanisms by which parasitic wasps manipulate host lipid homeostasis and may help to expand our knowledge of other parasitic systems.
Collapse
Affiliation(s)
- Yanping Wang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Xiaotong Wu
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Zehua Wang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Ting Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Sicong Zhou
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Jiani Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Lan Pang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Xiqian Ye
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Min Shi
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
| | - Jianhua Huang
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
- * E-mail:
| | - Xuexin Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China
- Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insect Pests, Zhejiang University, Hangzhou, China
- Key Laboratory of Biology of Crop Pathogens and Insects of Zhejiang Province, Zhejiang University, Hangzhou, China
- State Key Lab of Rice Biology, Zhejiang University, Hangzhou, China
| |
Collapse
|
44
|
Ni L. Genetic Transsynaptic Techniques for Mapping Neural Circuits in Drosophila. Front Neural Circuits 2021; 15:749586. [PMID: 34675781 PMCID: PMC8524129 DOI: 10.3389/fncir.2021.749586] [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: 07/29/2021] [Accepted: 09/13/2021] [Indexed: 11/23/2022] Open
Abstract
A neural circuit is composed of a population of neurons that are interconnected by synapses and carry out a specific function when activated. It is the structural framework for all brain functions. Its impairments often cause diseases in the nervous system. To understand computations and functions in a brain circuit, it is of crucial importance to identify how neurons in this circuit are connected. Genetic transsynaptic techniques provide opportunities to efficiently answer this question. These techniques label synapses or across synapses to unbiasedly label synaptic partners. They allow for mapping neural circuits with high reproducibility and throughput, as well as provide genetic access to synaptically connected neurons that enables visualization and manipulation of these neurons simultaneously. This review focuses on three recently developed Drosophila genetic transsynaptic tools for detecting chemical synapses, highlights their advantages and potential pitfalls, and discusses the future development needs of these techniques.
Collapse
|
45
|
Dekmak AS, Yang X, Zu Dohna H, Buchon N, Osta MA. The Route of Infection Influences the Contribution of Key Immunity Genes to Antibacterial Defense in Anopheles gambiae. J Innate Immun 2020; 13:107-126. [PMID: 33207342 DOI: 10.1159/000511401] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
Insect systemic immune responses to bacterial infections have been mainly studied using microinjections, whereby the microbe is directly injected into the hemocoel. While this methodology has been instrumental in defining immune signaling pathways and enzymatic cascades in the hemolymph, it remains unclear whether and to what extent the contribution of systemic immune defenses to host microbial resistance varies if bacteria invade the hemolymph after crossing the midgut epithelium subsequent to an oral infection. Here, we address this question using the pathogenic Serratia marcescens (Sm) DB11 strain to establish systemic infections of the malaria vector Anopheles gambiae, either by septic Sm injections or by midgut crossing after feeding on Sm. Using functional genetic studies by RNAi, we report that the two humoral immune factors, thioester-containing protein 1 and C-type lectin 4, which play key roles in defense against Gram-negative bacterial infections, are essential for defense against systemic Sm infections established through injection, but they become dispensable when Sm infects the hemolymph following oral infection. Similar results were observed for the mosquito Rel2 pathway. Surprisingly, blocking phagocytosis by cytochalasin D treatment did not affect mosquito susceptibility to Sm infections established through either route. Transcriptomic analysis of mosquito midguts and abdomens by RNA-seq revealed that the transcriptional response in these tissues is more pronounced in response to feeding on Sm. Functional classification of differentially expressed transcripts identified metabolic genes as the most represented class in response to both routes of infection, while immune genes were poorly regulated in both routes. We also report that Sm oral infections are associated with significant downregulation of several immune genes belonging to different families, specifically the clip-domain serine protease family. In sum, our findings reveal that the route of infection not only alters the contribution of key immunity genes to host antimicrobial defense but is also associated with different transcriptional responses in midguts and abdomens, possibly reflecting different adaptive strategies of the host.
Collapse
Affiliation(s)
- Amira San Dekmak
- Biology Department, American University of Beirut, Beirut, Lebanon
| | - Xiaowei Yang
- Entomology Department, Cornell Institute for Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| | | | - Nicolas Buchon
- Entomology Department, Cornell Institute for Host-Microbe Interactions and Disease, Cornell University, Ithaca, New York, USA
| | - Mike A Osta
- Biology Department, American University of Beirut, Beirut, Lebanon,
| |
Collapse
|
46
|
Hadjieconomou D, King G, Gaspar P, Mineo A, Blackie L, Ameku T, Studd C, de Mendoza A, Diao F, White BH, Brown AEX, Plaçais PY, Préat T, Miguel-Aliaga I. Enteric neurons increase maternal food intake during reproduction. Nature 2020; 587:455-459. [PMID: 33116314 PMCID: PMC7610780 DOI: 10.1038/s41586-020-2866-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/04/2020] [Indexed: 01/01/2023]
Abstract
Reproduction induces increased food intake across females of many animal species1-4, providing a physiologically relevant paradigm for the exploration of appetite regulation. Here, by examining the diversity of enteric neurons in Drosophila melanogaster, we identify a key role for gut-innervating neurons with sex- and reproductive state-specific activity in sustaining the increased food intake of mothers during reproduction. Steroid and enteroendocrine hormones functionally remodel these neurons, which leads to the release of their neuropeptide onto the muscles of the crop-a stomach-like organ-after mating. Neuropeptide release changes the dynamics of crop enlargement, resulting in increased food intake, and preventing the post-mating remodelling of enteric neurons reduces both reproductive hyperphagia and reproductive fitness. The plasticity of enteric neurons is therefore key to reproductive success. Our findings provide a mechanism to attain the positive energy balance that sustains gestation, dysregulation of which could contribute to infertility or weight gain.
Collapse
Affiliation(s)
- Dafni Hadjieconomou
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - George King
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Pedro Gaspar
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Alessandro Mineo
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Laura Blackie
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Tomotsune Ameku
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Chris Studd
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Alex de Mendoza
- Australian Research Council Centre of Excellence in Plant Energy Biology, School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Fengqiu Diao
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin H White
- Laboratory of Molecular Biology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - André E X Brown
- MRC London Institute of Medical Sciences, London, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Pierre-Yves Plaçais
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Thomas Préat
- Genes and Dynamics of Memory Systems, Brain Plasticity Unit, CNRS, ESPCI Paris, PSL Research University, Paris, France
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, London, UK.
- Faculty of Medicine, Imperial College London, London, UK.
| |
Collapse
|
47
|
Koyama T, Texada MJ, Halberg KA, Rewitz K. Metabolism and growth adaptation to environmental conditions in Drosophila. Cell Mol Life Sci 2020; 77:4523-4551. [PMID: 32448994 PMCID: PMC7599194 DOI: 10.1007/s00018-020-03547-2] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 04/19/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
Organisms adapt to changing environments by adjusting their development, metabolism, and behavior to improve their chances of survival and reproduction. To achieve such flexibility, organisms must be able to sense and respond to changes in external environmental conditions and their internal state. Metabolic adaptation in response to altered nutrient availability is key to maintaining energy homeostasis and sustaining developmental growth. Furthermore, environmental variables exert major influences on growth and final adult body size in animals. This developmental plasticity depends on adaptive responses to internal state and external cues that are essential for developmental processes. Genetic studies have shown that the fruit fly Drosophila, similarly to mammals, regulates its metabolism, growth, and behavior in response to the environment through several key hormones including insulin, peptides with glucagon-like function, and steroid hormones. Here we review emerging evidence showing that various environmental cues and internal conditions are sensed in different organs that, via inter-organ communication, relay information to neuroendocrine centers that control insulin and steroid signaling. This review focuses on endocrine regulation of development, metabolism, and behavior in Drosophila, highlighting recent advances in the role of the neuroendocrine system as a signaling hub that integrates environmental inputs and drives adaptive responses.
Collapse
Affiliation(s)
- Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth A Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
48
|
Abstract
Abstract
Background
Organisms show an incredibly diverse array of body and organ shapes that are both unique to their taxon and important for adapting to their environment. Achieving these specific shapes involves coordinating the many processes that transform single cells into complex organs, and regulating their growth so that they can function within a fully-formed body.
Main text
Conceptually, body and organ shape can be separated in two categories, although in practice these categories need not be mutually exclusive. Body shape results from the extent to which organs, or parts of organs, grow relative to each other. The patterns of relative organ size are characterized using allometry. Organ shape, on the other hand, is defined as the geometric features of an organ’s component parts excluding its size. Characterization of organ shape is frequently described by the relative position of homologous features, known as landmarks, distributed throughout the organ. These descriptions fall into the domain of geometric morphometrics.
Conclusion
In this review, we discuss the methods of characterizing body and organ shape, the developmental programs thought to underlie each, highlight when and how the mechanisms regulating body and organ shape might overlap, and provide our perspective on future avenues of research.
Collapse
|
49
|
Zhao X, Karpac J. The Drosophila midgut and the systemic coordination of lipid-dependent energy homeostasis. CURRENT OPINION IN INSECT SCIENCE 2020; 41:100-105. [PMID: 32898765 PMCID: PMC7669600 DOI: 10.1016/j.cois.2020.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/19/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
The evolution of complex organ systems in metazoans has dictated that the maintenance of energy homeostasis requires coordinating local and systemic energy demands between organs with specialized functions. The gastrointestinal tract is one of many organs that is indispensable for the systemic coordination of energy substrate uptake, storage, and usage, and the spatial organization of this organ (i.e. proximity to other metabolic organs) within a complex body plan underlies its role in organ crosstalk. Studies of various arthropod intestines, and in particular insects, have shed light on the evolution and function of the gastrointestinal tract in the maintenance of energy homeostasis. This brief review focuses on studies and theories derived from the insect intestine (particularly the midgut) of adult Drosophila melanogaster to inform on the how, what, and why of the gastrointestinal tract in the systemic regulation of lipids, the most common form of stored energy in insects.
Collapse
Affiliation(s)
- Xiao Zhao
- Dept. of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA
| | - Jason Karpac
- Dept. of Molecular and Cellular Medicine, Texas A&M University Health Science Center, Bryan, TX 77807, USA.
| |
Collapse
|
50
|
Jiang SH, Zhang XX, Hu LP, Wang X, Li Q, Zhang XL, Li J, Gu JR, Zhang ZG. Systemic Regulation of Cancer Development by Neuro-Endocrine-Immune Signaling Network at Multiple Levels. Front Cell Dev Biol 2020; 8:586757. [PMID: 33117814 PMCID: PMC7561376 DOI: 10.3389/fcell.2020.586757] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/14/2020] [Indexed: 12/19/2022] Open
Abstract
The overarching view of current tumor therapies simplifies cancer to a cell-biology problem in which neoplasms are caused solely by malignant cells and the exploration of carcinogenesis and tumor progression largely focuses on somatic mutations and other genetic abnormalities of cancer cells. The limited therapeutic response indicates that cancer is driven not only by endogenous oncogenic factors and reciprocal interactions within the tumor microenvironment, but also by complex systemic processes. Homeostasis is the fundamental premise of health, and is maintained by systemic regulation of neuro-endocrine-immune axis. Cancer is also a systemic disease that manifested by dysfunction of the nervous, endocrine, and immune systems. Multiple axes of regulation exist in cancer, including central-, organ-, and microenvironment-level manipulation. At each specific regulatory level, the tridirectional communication among the nervous, endocrine, and immune factors transmit flexible signaling to induce proliferation, invasion, reprogrammed metabolism, therapeutic resistance, and other malignant phenotypes of cancer cells, resulting in the extremely poor prognosis of this lethal disease. Understanding this coordinated signaling network will enable the development of new approaches for cancer treatment via behavioral and pharmacological interventions.
Collapse
Affiliation(s)
- Shu-Heng Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiao-Xin Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Li-Peng Hu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qing Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xue-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Ren Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|