1
|
Biswas P, Bako JA, Liston JB, Yu H, Wat LW, Miller CJ, Gordon MD, Huan T, Stanley M, Rideout EJ. Insulin/insulin-like growth factor signaling pathway promotes higher fat storage in Drosophila females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.623936. [PMID: 40342968 PMCID: PMC12060994 DOI: 10.1101/2024.11.18.623936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
In Drosophila , adult females store more fat than males. While the mechanisms that restrict body fat in males are becoming clearer, less is known about how females achieve higher fat storage. Here, we perform a detailed investigation of the mechanisms that promote higher fat storage in females. We show greater intake of dietary sugar supports higher fat storage due to female-biased remodeling of the fat body lipidome. Dietary sugar stimulates a female-specific increase in Drosophila insulin-like peptide 3 (Dilp3), which acts together with greater peripheral insulin sensitivity to augment insulin/insulin-like growth factor signaling pathway (IIS) activity in adult females. Indeed, Dilp3 overexpression prevented the female-biased decrease in body fat after removal of dietary sugar. Given that adult-specific IIS inhibition caused a female-biased decrease in body fat, our data reveal IIS as a key determinant of female fat storage.
Collapse
|
2
|
Liu Y, Dantas E, Ferrer M, Miao T, Qadiri M, Liu Y, Comjean A, Davidson EE, Perrier T, Ahmed T, Hu Y, Goncalves MD, Janowitz T, Perrimon N. Hepatic gluconeogenesis and PDK3 upregulation drive cancer cachexia in flies and mice. Nat Metab 2025; 7:823-841. [PMID: 40275022 PMCID: PMC12021660 DOI: 10.1038/s42255-025-01265-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 03/06/2025] [Indexed: 04/26/2025]
Abstract
Cachexia, a severe wasting syndrome characterized by tumour-induced metabolic dysregulation, is a leading cause of death in people with cancer, yet its underlying mechanisms remain poorly understood. Here we show that a longitudinal full-body single-nuclei-resolution transcriptome analysis in a Drosophila model of cancer cachexia captures interorgan dysregulations. Our study reveals that the tumour-secreted interleukin-like cytokine Upd3 induces fat-body expression of Pepck1 and Pdk, key regulators of gluconeogenesis, disrupting glucose metabolism and contributing to cachexia. Similarly, in mouse cancer cachexia models, we observe IL-6-JAK-STAT-signalling-mediated induction of Pck1 and Pdk3 expression in the liver. Increased expression of these genes in fly, mouse, and human correlates with poor prognosis, and hepatic expression of Pdk3 emerges as a previously unknown mechanism contributing to metabolic dysfunction in cancer cachexia. This study highlights the conserved nature of tumour-induced metabolic disruptions and identifies potential therapeutic targets to mitigate cachexia in people with cancer.
Collapse
Affiliation(s)
- Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Ezequiel Dantas
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Ting Miao
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Mujeeb Qadiri
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Emma E Davidson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Ohio State University College of Medicine, Columbus, OH, USA
| | - Tiffany Perrier
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Tanvir Ahmed
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Marcus D Goncalves
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, New York, NY, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
3
|
Sigurðardóttir H, Ablondi M, Kristjansson T, Lindgren G, Eriksson S. Genetic diversity and signatures of selection in Icelandic horses and Exmoor ponies. BMC Genomics 2024; 25:772. [PMID: 39118059 PMCID: PMC11308356 DOI: 10.1186/s12864-024-10682-8] [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: 03/27/2024] [Accepted: 08/01/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND The Icelandic horse and Exmoor pony are ancient, native breeds, adapted to harsh environmental conditions and they have both undergone severe historic bottlenecks. However, in modern days, the selection pressures on these breeds differ substantially. The aim of this study was to assess genetic diversity in both breeds through expected (HE) and observed heterozygosity (HO) and effective population size (Ne). Furthermore, we aimed to identify runs of homozygosity (ROH) to estimate and compare genomic inbreeding and signatures of selection in the breeds. RESULTS HO was estimated at 0.34 and 0.33 in the Icelandic horse and Exmoor pony, respectively, aligning closely with HE of 0.34 for both breeds. Based on genomic data, the Ne for the last generation was calculated to be 125 individuals for Icelandic horses and 42 for Exmoor ponies. Genomic inbreeding coefficient (FROH) ranged from 0.08 to 0.20 for the Icelandic horse and 0.12 to 0.27 for the Exmoor pony, with the majority of inbreeding attributed to short ROHs in both breeds. Several ROH islands associated with performance were identified in the Icelandic horse, featuring target genes such as DMRT3, DOCK8, EDNRB, SLAIN1, and NEURL1. Shared ROH islands between both breeds were linked to metabolic processes (FOXO1), body size, and the immune system (CYRIB), while private ROH islands in Exmoor ponies were associated with coat colours (ASIP, TBX3, OCA2), immune system (LYG1, LYG2), and fertility (TEX14, SPO11, ADAM20). CONCLUSIONS Evaluations of genetic diversity and inbreeding reveal insights into the evolutionary trajectories of both breeds, highlighting the consequences of population bottlenecks. While the genetic diversity in the Icelandic horse is acceptable, a critically low genetic diversity was estimated for the Exmoor pony, which requires further validation. Identified signatures of selection highlight the differences in the use of the two breeds as well as their adaptive trait similarities. The results provide insight into genomic regions under selection pressure in a gaited performance horse breed and various adaptive traits in small-sized native horse breeds. This understanding contributes to preserving genetic diversity and population health in these equine populations.
Collapse
Affiliation(s)
- Heiðrún Sigurðardóttir
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, 75007, Sweden.
- Faculty of Agricultural Sciences, Agricultural University of Iceland, Hvanneyri, Borgarbyggð, 311, Iceland.
| | - Michela Ablondi
- Department of Veterinary Science, University of Parma, Parma, 43126, Italy
| | - Thorvaldur Kristjansson
- Faculty of Agricultural Sciences, Agricultural University of Iceland, Hvanneyri, Borgarbyggð, 311, Iceland
| | - Gabriella Lindgren
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, 75007, Sweden
- Center for Animal Breeding and Genetics, Department of Biosystems, KU Leuven, Leuven, 3001, Belgium
| | - Susanne Eriksson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, P.O. Box 7023, Uppsala, 75007, Sweden
| |
Collapse
|
4
|
Montaser O, El-Aasr M, Tawfik HO, Meshrif WS, Elbrense H. Drosophila melanogaster as a model organism for diabetes II treatment by the ethyl acetate fraction of Atriplex halimus L. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2024; 341:702-716. [PMID: 38623920 DOI: 10.1002/jez.2812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/17/2024]
Abstract
Type 2 diabetes (T2D) is the most common metabolic disorder. The undesirable effects of synthetic drugs demand a search for safe antidiabetic agents. This study aimed to assess the antidiabetic activity of different fractions of Atriplex halimus (petroleum ether 60-80, methylene chloride, ethyl acetate, and n-butanol) using Drosophila melanogaster larvae. Titers of total glucose and trehalose, as well as larval weight, were measured and compared with those of control and diabetic larvae. The expression of Drosophila insulin-like peptides (DILP2 and DILP3) and adipokinetic hormone (AKH) was evaluated. The results revealed a significant increase in total glucose, trehalose, and a decrease in body weight in the larvae fed a high-sugar diet compared with those in the control. When larvae fed diets containing the tested fractions, the total glucose and trehalose decreased to the control level, and the body weight increased. DILP2, DILP3, and AKH exhibited significant decreases upon treatment with A. halimus ethyl acetate. Metabolomic profiling of the ethyl acetate fraction of A. halimus revealed the presence of flavonoids and flavonoid glycosides. After docking screening to predict the most powerful moiety, we discovered that flavonoid glycosides (especially eriodictyol-7-O-neohesperidoside) have a greater affinity for the pocket than the other moieties. The results indicated the therapeutic activity of the A. halimus ethyl acetate fraction against induced T2D in Drosophila larvae. The antidiabetic activity may be attributed to flavonoids, which are the main components of the A. halimus ethyl acetate fraction.
Collapse
Affiliation(s)
- Omnia Montaser
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Mona El-Aasr
- Department of Pharmacognosy, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Wesam S Meshrif
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| | - Hanaa Elbrense
- Department of Zoology, Faculty of Science, Tanta University, Tanta, Egypt
| |
Collapse
|
5
|
Leyria J. Endocrine factors modulating vitellogenesis and oogenesis in insects: An update. Mol Cell Endocrinol 2024; 587:112211. [PMID: 38494046 DOI: 10.1016/j.mce.2024.112211] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/26/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
The endocrine system plays a pivotal role in shaping the mechanisms that ensure successful reproduction. With over a million known insect species, understanding the endocrine control of reproduction has become increasingly complex. Some of the key players include the classic insect lipid hormones juvenile hormone (JH) and ecdysteroids, and neuropeptides such as insulin-like peptides (ILPs). Individual endocrine factors not only modulate their own target tissue but also play crucial roles in crosstalk among themselves, ensuring successful vitellogenesis and oogenesis. Recent advances in omics, gene silencing, and genome editing approaches have accelerated research, offering both fundamental insights and practical applications for studying in-depth endocrine signaling pathways. This review provides an updated and integrated view of endocrine factors modulating vitellogenesis and oogenesis in insect females.
Collapse
Affiliation(s)
- Jimena Leyria
- Department of Biology, University of Toronto Mississauga, Mississauga, ON, Canada.
| |
Collapse
|
6
|
Chao CF, Pesch YY, Yu H, Wang C, Aristizabal MJ, Huan T, Tanentzapf G, Rideout E. An important role for triglyceride in regulating spermatogenesis. eLife 2024; 12:RP87523. [PMID: 38805376 PMCID: PMC11132686 DOI: 10.7554/elife.87523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024] Open
Abstract
Drosophila is a powerful model to study how lipids affect spermatogenesis. Yet, the contribution of neutral lipids, a major lipid group which resides in organelles called lipid droplets (LD), to sperm development is largely unknown. Emerging evidence suggests LD are present in the testis and that loss of neutral lipid- and LD-associated genes causes subfertility; however, key regulators of testis neutral lipids and LD remain unclear. Here, we show LD are present in early-stage somatic and germline cells within the Drosophila testis. We identified a role for triglyceride lipase brummer (bmm) in regulating testis LD, and found that whole-body loss of bmm leads to defects in sperm development. Importantly, these represent cell-autonomous roles for bmm in regulating testis LD and spermatogenesis. Because lipidomic analysis of bmm mutants revealed excess triglyceride accumulation, and spermatogenic defects in bmm mutants were rescued by genetically blocking triglyceride synthesis, our data suggest that bmm-mediated regulation of triglyceride influences sperm development. This identifies triglyceride as an important neutral lipid that contributes to Drosophila sperm development, and reveals a key role for bmm in regulating testis triglyceride levels during spermatogenesis.
Collapse
Affiliation(s)
- Charlotte F Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Yanina-Yasmin Pesch
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Huaxu Yu
- Department of Chemistry, The University of British ColumbiaVancouverCanada
| | - Chenjingyi Wang
- Department of Chemistry, The University of British ColumbiaVancouverCanada
| | | | - Tao Huan
- Department of Chemistry, The University of British ColumbiaVancouverCanada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Elizabeth Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| |
Collapse
|
7
|
Cavieres-Lepe J, Amini E, Zabel M, Nässel DR, Stanewsky R, Wegener C, Ewer J. Timed receptor tyrosine kinase signaling couples the central and a peripheral circadian clock in Drosophila. Proc Natl Acad Sci U S A 2024; 121:e2308067121. [PMID: 38442160 PMCID: PMC10945756 DOI: 10.1073/pnas.2308067121] [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: 05/16/2023] [Accepted: 02/01/2024] [Indexed: 03/07/2024] Open
Abstract
Circadian clocks impose daily periodicities to behavior, physiology, and metabolism. This control is mediated by a central clock and by peripheral clocks, which are synchronized to provide the organism with a unified time through mechanisms that are not fully understood. Here, we characterized in Drosophila the cellular and molecular mechanisms involved in coupling the central clock and the peripheral clock located in the prothoracic gland (PG), which together control the circadian rhythm of emergence of adult flies. The time signal from central clock neurons is transmitted via small neuropeptide F (sNPF) to neurons that produce the neuropeptide Prothoracicotropic Hormone (PTTH), which is then translated into daily oscillations of Ca2+ concentration and PTTH levels. PTTH signaling is required at the end of metamorphosis and transmits time information to the PG through changes in the expression of the PTTH receptor tyrosine kinase (RTK), TORSO, and of ERK phosphorylation, a key component of PTTH transduction. In addition to PTTH, we demonstrate that signaling mediated by other RTKs contributes to the rhythmicity of emergence. Interestingly, the ligand to one of these receptors (Pvf2) plays an autocrine role in the PG, which may explain why both central brain and PG clocks are required for the circadian gating of emergence. Our findings show that the coupling between the central and the PG clock is unexpectedly complex and involves several RTKs that act in concert and could serve as a paradigm to understand how circadian clocks are coordinated.
Collapse
Affiliation(s)
- Javier Cavieres-Lepe
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso2360102, Chile
| | - Emad Amini
- Julius-Maximilians-Universität Würzburg, Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Am Hubland, Würzburg97074, Germany
| | - Maia Zabel
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso2360102, Chile
| | - Dick R. Nässel
- Department of Zoology, Stockholm University, 10691Stockholm, Sweden
| | - Ralf Stanewsky
- Institute of Neuro- and Behavioral Biology, Multiscale Imaging Centre, University of Münster, 48149Münster, Germany
| | - Christian Wegener
- Julius-Maximilians-Universität Würzburg, Biocenter, Theodor-Boveri-Institute, Neurobiology and Genetics, Am Hubland, Würzburg97074, Germany
| | - John Ewer
- Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso2360102, Chile
- Instituto de Neurociencias, Universidad de Valparaíso, Valparaíso2360102, Chile
| |
Collapse
|
8
|
Li X, Li W, Zhang S, Sang W, Peng Y, Zhao Y. RNA interference against the putative insulin receptor substrate gene IRS1 affects growth and development in the pest natural enemy Pardosa pseudoannulata. PEST MANAGEMENT SCIENCE 2024; 80:648-660. [PMID: 37756442 DOI: 10.1002/ps.7792] [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: 07/04/2023] [Revised: 08/29/2023] [Accepted: 09/27/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Insulin signalling pathways play crucial roles in regulating growth and development in insects, but their effects on the growth and development of Arachnids, such as spiders, have rarely been studied. As a valuable pest natural enemy in agricultural fields, the molecular mechanisms of insulin signalling pathway-mediated growth and development of the wolf spider, Pardosa pseudoannulata, are of particular interest. RESULTS In this study, we identified and characterized six insulin signalling pathway genes - InR, InR2, IRS1, PI3K1, PI3K2, and PDK - in Pardosa pseudoannulata. Real-time quantitative polymerase chain reaction results were used to analyse the relative expression levels of the six genes in different developmental instars and tissues, and in response to starvation treatment. In addition, the function of the insulin receptor substrate (IRS1) gene was investigated using RNA interference technology, which found that IRS1 significantly influenced nutrient content, developmental duration, body weight, and gonad development. CONCLUSION This study revealed the roles of six key insulin signalling pathway genes in Pardosa pseudoannulata, and in particular the importance of the IRS1 gene in regulating growth and development in the spider. The results lay the foundation for further research on the internal regulation mechanisms of growth and development in Araneae species, and also provide a reference for the artificial breeding of spiders. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xuelai Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Wei Li
- Hubei Key Laboratory of Regional Development and Environmental Response, Faculty of Resources and Environmental Science, Hubei University, Wuhan, China
| | - Shichang Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Wen Sang
- College of Plant Protection, South China Agricultural University, Guangzhou, China
| | - Yu Peng
- Hubei Key Laboratory of Regional Development and Environmental Response, Faculty of Resources and Environmental Science, Hubei University, Wuhan, China
| | - Yao Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
9
|
Voulgari-Kokota A, Boatta F, Rijkers R, Wertheim B, Beukeboom LW, Ellers J, Salles JF. High-sugar diet leads to loss of beneficial probiotics in housefly larvae guts. THE ISME JOURNAL 2024; 18:wrae193. [PMID: 39361901 PMCID: PMC11495414 DOI: 10.1093/ismejo/wrae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/15/2024] [Accepted: 10/01/2024] [Indexed: 10/05/2024]
Abstract
The housefly (Musca domestica) is a common insect species with only a few recurrent bacterial taxa in its gut microbiota, because the numerous microbial acquisition routes in its septic habitats can favor transient microbes. Here, we investigated the role of the diet on the microbiota and the developmental success of a housefly strain reared on three substrates. We used a control wheat bran-based substrate, and added clotted cream and sucrose to make a high-fat, and a high-sugar substrate, respectively. The conducted survey revealed that, in contrast to the high-fat diet, the high-sugar diet caused lower developmental success and less diverse microbiota, in which several lactobacilli were replaced with Weissella bacterial phylotypes. Cultures with sucrose as the sole carbon source confirmed that a Weissella confusa strain, isolated from larvae, could utilize sucrose more efficiently than other tested lactic acid bacteria; a result also supported by gene function prediction analysis. Enhancing the rearing substrate with Limosilactobacillus fermentum and Lactiplantibacillus plantarum strains, which were isolated from control larvae, could not only revert the negative effect of the high-sucrose diet on development, but also increase the gut bacterial diversity. In our study, we show that the microbiota shifts in response to the high-sucrose diet did not benefit the host, that showed lower developmental success. In contrast, high-sucrose favored specific components of the microbiota, that continued to be enriched even after multiple generations, outcompeting beneficial bacteria. Also, microbiome manipulation showed the potential of probiotics to rescue host performance and restore the microbiome.
Collapse
Affiliation(s)
- Anna Voulgari-Kokota
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, P.O. Box 11103, Groningen 9700 CC, The Netherlands
- Laboratory of Microbiology, Wageningen University, Wageningen 6700 EH, The Netherlands
| | - Francesco Boatta
- Amsterdam Institute for Life and Environment, Section Ecology and Evolution, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Ruud Rijkers
- Amsterdam Institute for Life and Environment, Section Ecology and Evolution, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
- Department of Environmental Science, Stockholm University, Stockholm SE-106 91, Sweden
| | - Bregje Wertheim
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, P.O. Box 11103, Groningen 9700 CC, The Netherlands
| | - Leo W Beukeboom
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, P.O. Box 11103, Groningen 9700 CC, The Netherlands
| | - Jacintha Ellers
- Laboratory of Microbiology, Wageningen University, Wageningen 6700 EH, The Netherlands
| | - Joana Falcao Salles
- Groningen Institute for Evolutionary Life Sciences (GELIFES), University of Groningen, Nijenborgh 7, P.O. Box 11103, Groningen 9700 CC, The Netherlands
| |
Collapse
|
10
|
Huang WH, Kajal K, Wibowo RH, Amartuvshin O, Kao SH, Rastegari E, Lin CH, Chiou KL, Pi HW, Ting CT, Hsu HJ. Excess dietary sugar impairs Drosophila adult stem cells via elevated reactive oxygen species-induced JNK signaling. Development 2024; 151:dev201772. [PMID: 38063853 DOI: 10.1242/dev.201772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 11/24/2023] [Indexed: 01/03/2024]
Abstract
High-sugar diets (HSDs) often lead to obesity and type 2 diabetes, both metabolic syndromes associated with stem cell dysfunction. However, it is unclear whether excess dietary sugar affects stem cells. Here, we report that HSD impairs stem cell function in the intestine and ovaries of female Drosophila prior to the onset of insulin resistance, a hallmark of type 2 diabetes. Although 1 week of HSD leads to obesity, impaired oogenesis and altered lipid metabolism, insulin resistance does not occur. HSD increases glucose uptake by germline stem cells (GSCs) and triggers reactive oxygen species-induced JNK signaling, which reduces GSC proliferation. Removal of excess sugar from the diet reverses these HSD-induced phenomena. A similar phenomenon is found in intestinal stem cells (ISCs), except that HSD disrupts ISC maintenance and differentiation. Interestingly, tumor-like GSCs and ISCs are less responsive to HSD, which may be because of their dependence on glycolytic metabolism and high energy demand, respectively. This study suggests that excess dietary sugar induces oxidative stress and damages stem cells before insulin resistance develops, a mechanism that may also occur in higher organisms.
Collapse
Affiliation(s)
- Wei-Hao Huang
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Department of Life Science, National Taiwan University, Taipei 10917
| | - Kreeti Kajal
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Molecular and Biological Agricultural Sciences Program, Taiwan International Graduate Program, National Chung Hsing University and Academia Sinica, Taipei 11529
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung 40227
| | | | - Oyundari Amartuvshin
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei 11529
- Graduate Institute of Life Science, National Defense Medical Center, Taipei 11490
| | - Shih-Han Kao
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
| | - Elham Rastegari
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
| | - Chi-Hung Lin
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica, Taipei 11529
- Graduate Institute of Life Science, National Defense Medical Center, Taipei 11490
| | - Kuan-Lin Chiou
- Department of Biomedical Science, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Hai-Wei Pi
- Department of Biomedical Science, College of Medicine, Chang Gung University, Tao-Yuan 333, Taiwan
| | - Chau-Ti Ting
- Department of Life Science, National Taiwan University, Taipei 10917
| | - Hwei-Jan Hsu
- Institute of Cellular and Organismic Biology, Sinica, Taipei 11529
| |
Collapse
|
11
|
Thompson KD, Suber W, Nicholas R, Arnosti DN. Long-range repression by ecdysone receptor on complex enhancers of the insulin receptor gene. Fly (Austin) 2023; 17:2242238. [PMID: 37621079 PMCID: PMC10461493 DOI: 10.1080/19336934.2023.2242238] [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: 06/16/2023] [Revised: 07/17/2023] [Accepted: 07/19/2023] [Indexed: 08/26/2023] Open
Abstract
The insulin signalling pathway is evolutionarily conserved throughout metazoans, playing key roles in development, growth, and metabolism. Misregulation of this pathway is associated with a multitude of disease states including diabetes, cancer, and neurodegeneration. The human insulin receptor gene (INSR) is widely expressed throughout development and was previously described as a 'housekeeping' gene. Yet, there is abundant evidence that this gene is expressed in a cell-type specific manner, with dynamic regulation in response to environmental signals. The Drosophila insulin-like receptor gene (InR) is homologous to the human INSR gene and was previously shown to be regulated by multiple transcriptional elements located primarily within the introns of the gene. These elements were roughly defined in ~1.5 kbp segments, but we lack an understanding of the potential detailed mechanisms of their regulation. We characterized the substructure of these cis-regulatory elements in Drosophila S2 cells, focusing on regulation through the ecdysone receptor (EcR) and the dFOXO transcription factor. By identifying specific locations of activators and repressors within 300 bp subelements, we show that some previously identified enhancers consist of relatively compact clusters of activators, while others have a distributed architecture not amenable to further reduction. In addition, these assays uncovered a long-range repressive action of unliganded EcR. The complex transcriptional circuitry likely endows InR with a highly flexible and tissue-specific response to tune insulin signalling. Further studies will provide insights to demonstrate the impact of natural variation in this gene's regulation, applicable to human genetic studies.
Collapse
Affiliation(s)
- Katie D. Thompson
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Will Suber
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, USA
| | - Rachel Nicholas
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - David N. Arnosti
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
12
|
Li Y, Pan L, Li P, Gao F, Wang L, Chen J, Li Z, Gao Y, Gong Y, Jin F. Isolation of Enterococcus faecium and determination of its mechanism for promoting the growth and development of Drosophila. Sci Rep 2023; 13:18726. [PMID: 37907538 PMCID: PMC10618532 DOI: 10.1038/s41598-023-43727-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 09/27/2023] [Indexed: 11/02/2023] Open
Abstract
Intestinal symbiotic microorganisms have a strong capacity to regulate the physiological functions of their host, and Drosophila serves as a useful model. Enterococcus faecium (E. faecium) is a member of the normal intestinal flora of animals. Lactic acid bacteria (LAB) such as E. faecium can promote the growth and development of Drosophila, but the mechanism of regulation of Drosophila is poorly understood. In this study, we found that E. faecium used a carbon source to produce probiotic acids. E. faecium is a symbiotic bacterium for Drosophila, and adult flies passed on parental flora to offspring. E. faecium promoted the growth and development of Drosophila, especially under poor nutritional conditions. E. faecium shortened the developmental process for Drosophila and accelerated the transformation from larva to pupa. Finally, E. faecium promoted the growth and development of Drosophila through TOR and insulin signalling pathways.
Collapse
Affiliation(s)
- Yujuan Li
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Lei Pan
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Pengcheng Li
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Fuguo Gao
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Lei Wang
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Jian Chen
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Zhichao Li
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China
| | - Yongheng Gao
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China.
| | - Yumei Gong
- Department of Surgery, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
| | - Faguang Jin
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital of Air Force Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
13
|
Li Y, Chang P, Sankaran S, Jang H, Nie Y, Zeng A, Hussain S, Wu JY, Chen X, Shi L. Bioorthogonal Stimulated Raman Scattering Imaging Uncovers Lipid Metabolic Dynamics in Drosophila Brain During Aging. GEN BIOTECHNOLOGY 2023; 2:247-261. [PMID: 37363411 PMCID: PMC10286263 DOI: 10.1089/genbio.2023.0017] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 05/31/2023] [Indexed: 06/28/2023]
Abstract
Studies have shown that brain lipid metabolism is associated with biological aging and influenced by dietary and genetic manipulations; however, the underlying mechanisms are elusive. High-resolution imaging techniques propose a novel and potent approach to understanding lipid metabolic dynamics in situ. Applying deuterium water (D2O) probing with stimulated Raman scattering (DO-SRS) microscopy, we revealed that lipid metabolic activity in Drosophila brain decreased with aging in a sex-dependent manner. Female flies showed an earlier occurrence of lipid turnover decrease than males. Dietary restriction (DR) and downregulation of insulin/IGF-1 signaling (IIS) pathway, two scenarios for lifespan extension, led to significant enhancements of brain lipid turnover in old flies. Combining SRS imaging with deuterated bioorthogonal probes (deuterated glucose and deuterated acetate), we discovered that, under DR treatment and downregulation of IIS pathway, brain metabolism shifted to use acetate as a major carbon source for lipid synthesis. For the first time, our study directly visualizes and quantifies spatiotemporal alterations of lipid turnover in Drosophila brain at the single organelle (lipid droplet) level. Our study not only demonstrates a new approach for studying brain lipid metabolic activity in situ but also illuminates the interconnection of aging, dietary, and genetic manipulations on brain lipid metabolic regulation.
Collapse
Affiliation(s)
- Yajuan Li
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Phyllis Chang
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Shriya Sankaran
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Hongje Jang
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Yuhang Nie
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Audrey Zeng
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Sahran Hussain
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Jane Y. Wu
- Department of Neurology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Xu Chen
- Department of Neurosciences, School of Medicine, University of California San Diego, La Jolla, California, USA
| | - Lingyan Shi
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
14
|
Thompson K, Suber W, Nicholas R, Arnosti DN. Long-range repression by ecdysone receptor on complex enhancers of the insulin receptor gene. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.23.541945. [PMID: 37293119 PMCID: PMC10245858 DOI: 10.1101/2023.05.23.541945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The insulin signaling pathway is evolutionarily conserved throughout metazoans, playing key roles in development, growth, and metabolism. Misregulation of this pathway is associated with a multitude of disease states including diabetes, cancer, and neurodegeneration. Genome-wide association studies indicate that natural variants in putative intronic regulatory elements of the human insulin receptor gene ( INSR) are associated with metabolic conditions, however, this gene's transcriptional regulation remains incompletely studied. INSR is widely expressed throughout development and was previously described as a 'housekeeping' gene. Yet, there is abundant evidence that this gene is expressed in a cell-type specific manner, with dynamic regulation in response to environmental signals. The Drosophila insulin-like receptor gene ( InR ) is homologous to the human INSR gene and was previously shown to be regulated by multiple transcriptional elements located primarily within the introns of the gene. These elements were roughly defined in ∼1.5 kbp segments, but we lack an understanding of the potential detailed mechanisms of their regulation, as well as the integrative output of the battery of enhancers in the entire locus. Using luciferase assays, we characterized the substructure of these cis-regulatory elements in Drosophila S2 cells, focusing on regulation through the ecdysone receptor (EcR) and the dFOXO transcription factor. The direct action of EcR on Enhancer 2 reveals a bimodal form of regulation, with active repression in the absence of the ligand, and positive activation in the presence of 20E. By identifying the location of activators of this enhancer, we characterized a long-range of repression acting over at least 475 bp, similar to the action of long-range repressors found in the embryo. dFOXO and 20E have contrasting effects on some of the individual regulatory elements, and for the adjacent enhancers 2 and 3, their influence was/was not found to be additive, indicating that enhancer action on this locus can/cannot be characterized in part by additive models. Other characterized enhancers from within this locus exhibited "distributed" or "localized" modes of action, suggesting that predicting the joint functional output of multiple regulatory regions will require a deeper experimental characterization. The noncoding intronic regions of InR have demonstrated dynamic regulation of expression and cell type specificity. This complex transcriptional circuitry goes beyond the simple conception of a 'housekeeping' gene. Further studies are aimed at identifying how these elements work together in vivo to generate finely tuned expression in tissue- and temporal-specific manners, to provide a guide to understanding the impact of natural variation in this gene's regulation, applicable to human genetic studies.
Collapse
Affiliation(s)
- Katie Thompson
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - Will Suber
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, Michigan
| | - Rachel Nicholas
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| | - David N Arnosti
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
15
|
Liu Y, Dantas E, Ferrer M, Liu Y, Comjean A, Davidson EE, Hu Y, Goncalves MD, Janowitz T, Perrimon N. Tumor Cytokine-Induced Hepatic Gluconeogenesis Contributes to Cancer Cachexia: Insights from Full Body Single Nuclei Sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540823. [PMID: 37292804 PMCID: PMC10245574 DOI: 10.1101/2023.05.15.540823] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A primary cause of death in cancer patients is cachexia, a wasting syndrome attributed to tumor-induced metabolic dysregulation. Despite the major impact of cachexia on the treatment, quality of life, and survival of cancer patients, relatively little is known about the underlying pathogenic mechanisms. Hyperglycemia detected in glucose tolerance test is one of the earliest metabolic abnormalities observed in cancer patients; however, the pathogenesis by which tumors influence blood sugar levels remains poorly understood. Here, utilizing a Drosophila model, we demonstrate that the tumor secreted interleukin-like cytokine Upd3 induces fat body expression of Pepck1 and Pdk, two key regulatory enzymes of gluconeogenesis, contributing to hyperglycemia. Our data further indicate a conserved regulation of these genes by IL-6/JAK-STAT signaling in mouse models. Importantly, in both fly and mouse cancer cachexia models, elevated gluconeogenesis gene levels are associated with poor prognosis. Altogether, our study uncovers a conserved role of Upd3/IL-6/JAK-STAT signaling in inducing tumor-associated hyperglycemia, which provides insights into the pathogenesis of IL-6 signaling in cancer cachexia.
Collapse
Affiliation(s)
- Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724 USA
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Emma E. Davidson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724 USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Marcus D. Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724 USA
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, New York, NY 11042 USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
16
|
Gao Y, Yang L, Chen Y, Liu P, Zhou Y, Chen X, Gu J. Aal-circRNA-407 regulates ovarian development of Aedes albopictus, a major arbovirus vector, via the miR-9a-5p/Foxl axis. PLoS Pathog 2023; 19:e1011374. [PMID: 37146060 DOI: 10.1371/journal.ppat.1011374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 05/17/2023] [Accepted: 04/19/2023] [Indexed: 05/07/2023] Open
Abstract
Aedes albopictus shows a rapid global expansion and dramatic vectorial capacity for various arboviruses, thus posing a severe threat to global health. Although many noncoding RNAs have been confirmed to play functional roles in various biological processes in Ae. albopictus, the roles of circRNA remain a mystery. In the present study, we first performed high-throughput circRNA sequencing in Ae. albopictus. Then, we identified a cysteine desulfurase (CsdA) superfamily gene-originated circRNA, named aal-circRNA-407, which was the third most abundant circRNA in adult females and displayed a fat body highly expressed manifestation and blood feeding-dependent onset. SiRNA-mediated knockdown of circRNA-407 resulted in a decrease in the number of developing follicles and a reduction in follicle size post blood meal. Furthermore, we demonstrated that circRNA-407 can act as a sponge of aal-miR-9a-5p to promote the expression of its target gene Foxl and eventually regulate ovarian development. Our study is the first to report a functional circRNA in mosquitoes, expanding our current understanding of important biological roles in mosquitoes and providing an alternative genetic strategy for mosquito control.
Collapse
Affiliation(s)
- Yonghui Gao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Lu Yang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Yulan Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Peiwen Liu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Ying Zhou
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoguang Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Jinbao Gu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Yoon KJ, Cunningham CB, Bretman A, Duncan EJ. One genome, multiple phenotypes: decoding the evolution and mechanisms of environmentally induced developmental plasticity in insects. Biochem Soc Trans 2023; 51:675-689. [PMID: 36929376 PMCID: PMC10246940 DOI: 10.1042/bst20210995] [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: 10/24/2022] [Revised: 02/16/2023] [Accepted: 02/21/2023] [Indexed: 03/18/2023]
Abstract
Plasticity in developmental processes gives rise to remarkable environmentally induced phenotypes. Some of the most striking and well-studied examples of developmental plasticity are seen in insects. For example, beetle horn size responds to nutritional state, butterfly eyespots are enlarged in response to temperature and humidity, and environmental cues also give rise to the queen and worker castes of eusocial insects. These phenotypes arise from essentially identical genomes in response to an environmental cue during development. Developmental plasticity is taxonomically widespread, affects individual fitness, and may act as a rapid-response mechanism allowing individuals to adapt to changing environments. Despite the importance and prevalence of developmental plasticity, there remains scant mechanistic understanding of how it works or evolves. In this review, we use key examples to discuss what is known about developmental plasticity in insects and identify fundamental gaps in the current knowledge. We highlight the importance of working towards a fully integrated understanding of developmental plasticity in a diverse range of species. Furthermore, we advocate for the use of comparative studies in an evo-devo framework to address how developmental plasticity works and how it evolves.
Collapse
Affiliation(s)
- Kane J. Yoon
- School of Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, U.K
| | | | - Amanda Bretman
- School of Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, U.K
| | - Elizabeth J. Duncan
- School of Biology, Faculty of Biological Sciences, University of Leeds, LS2 9JT Leeds, U.K
| |
Collapse
|
18
|
Gao Y, Cheng X, Tian Y, Yuan Z, Fan X, Yang D, Yang M. Nutritional Programming of the Lifespan of Male Drosophila by Activating FOXO on Larval Low-Nutrient Diet. Nutrients 2023; 15:nu15081840. [PMID: 37111059 PMCID: PMC10142539 DOI: 10.3390/nu15081840] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/30/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Nutrition during the developmental stages has long-term effects on adult physiology, disease and lifespan, and is termed nutritional programming. However, the underlying molecular mechanisms of nutritional programming are not yet well understood. In this study, we showed that developmental diets could regulate the lifespan of adult Drosophila in a way that interacts with various adult diets during development and adulthood. Importantly, we demonstrated that a developmental low-yeast diet (0.2SY) extended both the health span and lifespan of male flies under nutrient-replete conditions in adulthood through nutritional programming. Males with a low-yeast diets during developmental stages had a better resistance to starvation and lessened decline of climbing ability with age in adulthood. Critically, we revealed that the activity of the Drosophila transcription factor FOXO (dFOXO) was upregulated in adult males under developmental low-nutrient conditions. The knockdown of dFOXO, with both ubiquitous and fat-body-specific patterns, can completely abolish the lifespan-extending effect from the larval low-yeast diet. Ultimately, we identify that the developmental diet achieved the nutritional programming of the lifespan of adult males by modulating the activity of dFOXO in Drosophila. Together, these results provide molecular evidence that the nutrition in the early life of animals could program the health of their later life and their longevity.
Collapse
Affiliation(s)
- Yue Gao
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
| | - Xingyi Cheng
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
| | - Yao Tian
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
| | - Zhixiao Yuan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiaolan Fan
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Deying Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| | - Mingyao Yang
- Institute of Animal Genetics and Breeding, Sichuan Agricultural University, Chengdu 611130, China
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
19
|
Meng Q, Xu Y, Li Y, Wang Y. Novel studies on Drosophila melanogaster model reveal the roles of JNK-Jak/STAT axis and intestinal microbiota in insulin resistance. J Drug Target 2023; 31:261-268. [PMID: 36343203 DOI: 10.1080/1061186x.2022.2144869] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The JNK pathway play a critical role in insulin resistance induced by a long-term high-sugar diet. However, the roles of up- and downstream molecules of the JNK pathway in insulin resistance are less known in vertebrates and invertebrates. As a classical organism in biological research, Drosophila melanogaster (D. melanogaster) has been widely applied to the studies of mechanism of insulin resistance. Based on previous studies, we found a novel predictive mechanism of the formation of insulin resistance in D. melanogaster. We found that JNK activated by high-sugar diet and dysregulated intestinal microbiota could mediate inflammation, and then the activated JNK released Upd3, which in turn stimulated Jak/STAT pathway to release ImpL2. ImpL2 can compete with Drosophila insulin-like peptides (Dilps) for binding with the insulin receptor and inhibit the activation of insulin pathway. In this study, we reviewed novel studies on the insulin signalling pathway based on the D. melanogaster model. The findings support our hypothesis. We, therefore, described how a long-term high-sugar diet disrupts intestinal microbiota to induce inflammation and the disruption of JNK-Jak/STAT axis. This description may offer some new clues to the formation of insulin resistance.
Collapse
Affiliation(s)
- Qinghao Meng
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yidong Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Ying Li
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
20
|
Lovegrove MR, Dearden PK, Duncan EJ. Honeybee queen mandibular pheromone induces a starvation response in Drosophila melanogaster. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 154:103908. [PMID: 36657589 DOI: 10.1016/j.ibmb.2023.103908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 12/27/2022] [Accepted: 01/13/2023] [Indexed: 06/17/2023]
Abstract
Eusocial insect societies are defined by the reproductive division of labour, a social structure that is generally enforced by the reproductive dominant(s) or 'queen(s)'. Reproductive dominance is maintained through behavioural dominance or production of queen pheromones, or a mixture of both. Queen mandibular pheromone (QMP) is a queen pheromone produced by queen honeybees (Apis mellifera) which represses reproduction in worker honeybees. How QMP acts to repress worker reproduction, the mechanisms by which this repression is induced, and how it has evolved this activity, remain poorly understood. Surprisingly, QMP is capable of repressing reproduction in non-target arthropods. Here we show that in Drosophila melanogaster QMP treatment mimics the starvation response, disrupting reproduction. QMP exposure induces an increase in food consumption and activation of checkpoints in the ovary that reduce fecundity and depresses insulin signalling. The magnitude of these effects is indistinguishable between QMP-treated and starved individuals. As QMP triggers a starvation response in an insect diverged from honeybees, we propose that QMP originally evolved by co-opting nutrition signalling pathways to regulate reproduction.
Collapse
Affiliation(s)
- Mackenzie R Lovegrove
- Genomics Aotearoa and Biochemistry Department, University of Otago, P.O. Box 56, Dunedin, Aotearoa, New Zealand; School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Peter K Dearden
- Genomics Aotearoa and Biochemistry Department, University of Otago, P.O. Box 56, Dunedin, Aotearoa, New Zealand.
| | - Elizabeth J Duncan
- School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
21
|
Kim CY, Kim YG. Insulin-like Peptides of the Western Flower Thrips Frankliniella occidentalis and Their Mediation of Immature Development. INSECTS 2023; 14:47. [PMID: 36661977 PMCID: PMC9864108 DOI: 10.3390/insects14010047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 12/31/2022] [Accepted: 01/01/2023] [Indexed: 05/23/2023]
Abstract
Insulin-like peptides (ILPs) mediate various physiological processes in insects. Specifically, ILP expression is required for immature development in different insects. The western flower thrips, Frankliniella occidentalis, is polyphagous, but its occurrence and population density vary among different hosts. This study assesses the developmental variations in the thrips through quantitative analysis of their ILP expressions. Two types of ILPs (Fo-ILP1 and Fo-ILP2) were identified from the genome of F. occidentalis, and both ILPs were predicted to have the characteristics of signal peptides and B-C-A chains linked by cysteines. A phylogenetic analysis indicates that these two ILPs in the thrips are clustered with the ILP1 of Drosophila melanogaster, suggesting their physiological roles in growth. In addition, the two ILP genes were relatively highly expressed at all feeding stages, but their expression was reduced during the nonfeeding prepupal and pupal stages. Furthermore, RNA interference of each ILP expression led to significant developmental retardation. In validating the ILP expression in the thrips' development, five different varieties of host hot peppers were assessed in a choice test, along with the immature development of F. occidentalis. The expression levels of the two ILP genes were highly correlated with variations in the immature developmental rates of different hot pepper varieties. These suggest that Fo-ILP1 and Fo-ILP2 mediate the immature development of F. occidentalis by sensing different nutritional values of hot peppers. This study is the first report on ILPs in thysanopteran insects.
Collapse
Affiliation(s)
| | - Yong-Gyun Kim
- Department of Plant Medicals, College of Life Sciences, Andong National University, Andong 36729, Republic of Korea
| |
Collapse
|
22
|
Scalia P, Williams SJ, Fujita-Yamaguchi Y, Giordano A. Cell cycle control by the insulin-like growth factor signal: at the crossroad between cell growth and mitotic regulation. Cell Cycle 2023; 22:1-37. [PMID: 36005738 PMCID: PMC9769454 DOI: 10.1080/15384101.2022.2108117] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
In proliferating cells and tissues a number of checkpoints (G1/S and G2/M) preceding cell division (M-phase) require the signal provided by growth factors present in serum. IGFs (I and II) have been demonstrated to constitute key intrinsic components of the peptidic active fraction of mammalian serum. In vivo genetic ablation studies have shown that the cellular signal triggered by the IGFs through their cellular receptors represents a non-replaceable requirement for cell growth and cell cycle progression. Retroactive and current evaluation of published literature sheds light on the intracellular circuitry activated by these factors providing us with a better picture of the pleiotropic mechanistic actions by which IGFs regulate both cell size and mitogenesis under developmental growth as well as in malignant proliferation. The present work aims to summarize the cumulative knowledge learned from the IGF ligands/receptors and their intracellular signaling transducers towards control of cell size and cell-cycle with particular focus to their actionable circuits in human cancer. Furthermore, we bring novel perspectives on key functional discriminants of the IGF growth-mitogenic pathway allowing re-evaluation on some of its signal components based upon established evidences.
Collapse
Affiliation(s)
- Pierluigi Scalia
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,CST, Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United states,CONTACT Pierluigi Scalia ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA9102, USA
| | - Stephen J Williams
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,CST, Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, United states
| | - Yoko Fujita-Yamaguchi
- Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Antonio Giordano
- ISOPROG-Somatolink EPFP Research Network, Philadelphia, PA, USA, Caltanissetta, Italy,School of Medical Biotechnology, University of Siena, Italy
| |
Collapse
|
23
|
Donati Zeppa S, Agostini D, Ferrini F, Gervasi M, Barbieri E, Bartolacci A, Piccoli G, Saltarelli R, Sestili P, Stocchi V. Interventions on Gut Microbiota for Healthy Aging. Cells 2022; 12:cells12010034. [PMID: 36611827 PMCID: PMC9818603 DOI: 10.3390/cells12010034] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/15/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
In recent years, the improvement in health and social conditions has led to an increase in the average lifespan. Since aging is the most important risk factor for the majority of chronic human diseases, the development of therapies and intervention to stop, lessen or even reverse various age-related morbidities is an important target to ameliorate the quality of life of the elderly. The gut microbiota, that is, the complex ecosystem of microorganisms living in the gastrointestinal tract, plays an important role, not yet fully understood, in maintaining the host's health and homeostasis, influencing metabolic, oxidative and cognitive status; for this reason, it is also named "the forgotten endocrine organ" or "the second brain". On the other hand, the gut microbiota diversity and richness are affected by unmodifiable factors, such as aging and sex, and modifiable ones, such as diet, pharmacological therapies and lifestyle. In this review, we discuss the changes, mostly disadvantageous, for human health, induced by aging, in microbiota composition and the effects of dietary intervention, of supplementation with probiotics, prebiotics, synbiotics, psychobiotics and antioxidants and of physical exercise. The development of an integrated strategy to implement microbiota health will help in the goal of healthy aging.
Collapse
Affiliation(s)
- Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Deborah Agostini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Fabio Ferrini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence: (F.F.); (M.G.)
| | - Marco Gervasi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
- Correspondence: (F.F.); (M.G.)
| | - Elena Barbieri
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Alessia Bartolacci
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanni Piccoli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Roberta Saltarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Piero Sestili
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Vilberto Stocchi
- Department of Human Science for Promotion of Quality of Life, Univerity San Raffaele, 00166 Rome, Italy
| |
Collapse
|
24
|
Zhong S, Chèvre R, Castaño Mayan D, Corlianò M, Cochran BJ, Sem KP, van Dijk TH, Peng J, Tan LJ, Hartimath SV, Ramasamy B, Cheng P, Groen AK, Kuipers F, Goggi JL, Drum C, van Dam RM, Tan RS, Rye KA, Hayden MR, Cheng CY, Chacko S, Flannick J, Sim X, Tan HC, Singaraja RR. Haploinsufficiency of CYP8B1 associates with increased insulin sensitivity in humans. J Clin Invest 2022; 132:152961. [PMID: 36107630 PMCID: PMC9621133 DOI: 10.1172/jci152961] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/08/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUNDCytochrome P450 family 8 subfamily B member 1 (CYP8B1) generates 12α-hydroxylated bile acids (BAs) that are associated with insulin resistance in humans.METHODSTo determine whether reduced CYP8B1 activity improves insulin sensitivity, we sequenced CYP8B1 in individuals without diabetes and identified carriers of complete loss-of-function (CLOF) mutations utilizing functional assays.RESULTSMutation carriers had lower plasma 12α-hydroxylated/non-12α-hydroxylated BA and cholic acid (CA)/chenodeoxycholic acid (CDCA) ratios compared with age-, sex-, and BMI-matched controls. During insulin clamps, hepatic glucose production was suppressed to a similar magnitude by insulin, but glucose infusion rates to maintain euglycemia were higher in mutation carriers, indicating increased peripheral insulin sensitivity. Consistently, a polymorphic CLOF CYP8B1 mutation associated with lower fasting insulin in the AMP-T2D-GENES study. Exposure of primary human muscle cells to mutation-carrier CA/CDCA ratios demonstrated increased FOXO1 activity, and upregulation of both insulin signaling and glucose uptake, which were mediated by increased CDCA. Inhibition of FOXO1 attenuated the CDCA-mediated increase in muscle insulin signaling and glucose uptake. We found that reduced CYP8B1 activity associates with increased insulin sensitivity in humans.CONCLUSIONOur findings suggest that increased circulatory CDCA due to reduced CYP8B1 activity increases skeletal muscle insulin sensitivity, contributing to increased whole-body insulin sensitization.FUNDINGBiomedical Research Council/National Medical Research Council of Singapore.
Collapse
Affiliation(s)
- Shiqi Zhong
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Raphael Chèvre
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore
| | - David Castaño Mayan
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore.,Cardiovascular Research Institute, National University Health System, Singapore
| | - Maria Corlianò
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore.,Cardiovascular Research Institute, National University Health System, Singapore
| | - Blake J. Cochran
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Kai Ping Sem
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Theo H. van Dijk
- Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Netherlands
| | | | - Liang Juin Tan
- Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore
| | | | | | - Peter Cheng
- Singapore Bioimaging Consortium, A*STAR, Singapore
| | - Albert K. Groen
- Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Netherlands
| | - Folkert Kuipers
- Departments of Pediatrics and Laboratory Medicine, University of Groningen, University Medical Center Groningen, Netherlands
| | | | - Chester Drum
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Cardiovascular Research Institute, National University Health System, Singapore
| | - Rob M. van Dam
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Ru San Tan
- Department of Cardiology, National Heart Centre, Singapore
| | - Kerry-Anne Rye
- School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Michael R. Hayden
- Department of Medical Genetics, Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore.,Ophthalmology and Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore
| | - Shaji Chacko
- USDA/ARS Children’s Nutrition Research Centre, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Jason Flannick
- Program in Metabolism and,Program in Medical & Population Genetics, Broad Institute, Cambridge, Massachusetts, USA.,Division of Genetics and Genomics, Boston Children’s Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Xueling Sim
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, Singapore
| | - Hong Chang Tan
- Department of Endocrinology, Singapore General Hospital, Singapore
| | - Roshni R. Singaraja
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research (A*STAR), Singapore.,Cardiovascular Research Institute, National University Health System, Singapore
| |
Collapse
|
25
|
Kudla AM, Miranda X, Nijhout HF. The roles of growth regulation and appendage patterning genes in the morphogenesis of treehopper pronota. Proc Biol Sci 2022; 289:20212682. [PMID: 35673859 PMCID: PMC9174728 DOI: 10.1098/rspb.2021.2682] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Treehoppers of the insect family Membracidae have evolved enlarged and elaborate pronotal structures, which is hypothesized to involve co-opted expression of genes that are shared with the wings. Here, we investigate the similarity between the pronotum and wings in relation to growth. Our study reveals that the ontogenetic allometry of the pronotum is similar to that of wings in Membracidae, but not the outgroup. Using transcriptomics, we identify genes related to translation and protein synthesis, which are mutually upregulated. These genes are implicated in the eIF2, eIF4/p70S6K and mTOR pathways, and have known roles in regulating cell growth and proliferation. We find that species-specific differential growth patterning of the pronotum begins as early as the third instar, which suggests that expression of appendage patterning genes occurs long before the metamorphic molt. We propose that a network related to growth and size determination is the more likely mechanism shared with wings. However, regulators upstream of the shared genes in pronotum and wings need to be elucidated to substantiate whether co-option has occurred. Finally, we believe it will be helpful to distinguish the mechanisms leading to pronotal size from those regulating pronotal shape as we make sense of this spectacular evolutionary innovation.
Collapse
Affiliation(s)
- Anna M. Kudla
- Department of Biology, Duke University, Durham, NC 27708, USA
| | - Ximena Miranda
- Escuela de Biología, Universidad de Costa Rica, San José, Costa Rica
| | | |
Collapse
|
26
|
Chen J, Huang Y, Qi G. LncRNA-IRAR-mediated regulation of insulin receptor transcripts in Drosophila melanogaster during nutritional stress. INSECT MOLECULAR BIOLOGY 2022; 31:261-272. [PMID: 34923706 DOI: 10.1111/imb.12756] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/15/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
The insulin signalling pathway plays a crucial role in regulating the metabolism of sugars, fats and proteins in cells, thereby affecting the growth, metabolism, reproduction and ageing of organisms. However, little is known about the functions of long non-coding RNAs (lncRNAs) in the regulation of insulin receptors under stress conditions in insects. In this study, we showed that insulin receptor-associated lncRNA (IRAR) regulates insulin receptor transcripts in response to nutritional stress in Drosophila melanogaster. Genome editing by CRISPR-Cas9 showed reduced sensitivity of IRAR mutants to environmental nutritional changes. In contrast, the sensitivity of mutants overexpressing tubulin-gal4 > IRAR increased under low nutrition. The pupation and eclosion timings in IRAR mutants were significantly delayed with an increase in insulin concentration compared with that in the w1118 group. In addition, the expression pattern of IRAR was almost consistent with that of the four transcripts of the insulin receptor from the embryonic period to the adult period. RNA immunoprecipitation assay showed the direct regulation of insulin receptor transcripts by IRAR to the through FOXO binding under nutritional stress. To our knowledge, this is the first study that describes a model of lncRNA-mediated development regulation through insulin receptor transcripts.
Collapse
Affiliation(s)
- Jie Chen
- Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuantai Huang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guojun Qi
- Guangdong Provincial Key Laboratory of High Technology for Plant Protection, Plant Protection Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong, China
| |
Collapse
|
27
|
Millington JW, Biswas P, Chao C, Xia YH, Wat LW, Brownrigg GP, Sun Z, Basner-Collins PJ, Klein Geltink RI, Rideout EJ. A low-sugar diet enhances Drosophila body size in males and females via sex-specific mechanisms. Development 2022; 149:dev200491. [PMID: 35195254 PMCID: PMC10656461 DOI: 10.1242/dev.200491] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 02/14/2022] [Indexed: 12/11/2022]
Abstract
In Drosophila, changes to dietary protein elicit different body size responses between the sexes. Whether these differential body size effects extend to other macronutrients remains unclear. Here, we show that lowering dietary sugar (0S diet) enhanced body size in male and female larvae. Despite an equivalent phenotypic effect between the sexes, we detected sex-specific changes to signalling pathways, transcription and whole-body glycogen and protein. In males, the low-sugar diet augmented insulin/insulin-like growth factor signalling pathway (IIS) activity by increasing insulin sensitivity, where increased IIS was required for male metabolic and body size responses in 0S. In females reared on low sugar, IIS activity and insulin sensitivity were unaffected, and IIS function did not fully account for metabolic and body size responses. Instead, we identified a female-biased requirement for the Target of rapamycin pathway in regulating metabolic and body size responses. Together, our data suggest the mechanisms underlying the low-sugar-induced increase in body size are not fully shared between the sexes, highlighting the importance of including males and females in larval studies even when similar phenotypic outcomes are observed.
Collapse
Affiliation(s)
- Jason W. Millington
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Puja Biswas
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Charlotte Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Yi Han Xia
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Lianna W. Wat
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - George P. Brownrigg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Ziwei Sun
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Paige J. Basner-Collins
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| | - Ramon I. Klein Geltink
- Department of Pathology and Laboratory Medicine, British Columbia Children's Hospital Research Institute, Vancouver V5Z 4H4, Canada
| | - Elizabeth J. Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver V6T 1Z3, Canada
| |
Collapse
|
28
|
Cruz-Pineda WD, Garibay-Cerdenares OL, Rodríguez-Ruíz HA, Matia-García I, Marino-Ortega LA, Espinoza-Rojo M, Reyes-Castillo Z, Castro-Alarcón N, Castañeda-Saucedo E, Illades-Aguiar B, Parra-Rojas I. Changes in the Expression of Insulin Pathway, Neutrophil Elastase and Alpha 1 Antitrypsin Genes from Leukocytes of Young Individuals with Insulin Resistance. Diabetes Metab Syndr Obes 2022; 15:1865-1876. [PMID: 35757193 PMCID: PMC9215908 DOI: 10.2147/dmso.s362881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/19/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Chronic hyperinsulinemia is a hallmark of insulin resistance that affects a diversity of cells, including leukocytes modifying the expression of some genes involved in insulin signaling. PURPOSE The aim of this study was to evaluate how hyperinsulinemia affects the expression of genes involved in the proximal insulin signaling pathway in leukocytes from 45 young individuals grouped: normal weight with not insulin resistance (NIR), with insulin resistance (IR) and with obesity (OB-IR). METHODS qPCR was performed to analyze the expression of insulin receptor (INSR), insulin receptor substrate 1 and 2 (IRS-1 and IRS-2), neutrophil elastase (NE), alpha 1 antitrypsin (A1AT), glucose transporters 1, 3 and 4 (GLUT-1, GLUT-3 and GLUT-4) by the 2-ΔCt method, and the correlation between the genes was determined by Spearman's test. RESULTS The mRNA expression analysis of all genes between NIR and IR individuals revealed no differences. However, when comparing NIR and IR individuals with OB-IR, an increase in NE and A1AT expression and a clear trend towards a decrease in IRS-2 expression was observed, whereas the comparison of IR and OB-IR showed a decrease in GLUT-3 expression. Overall, the correlation analysis showed that in the IR group there was a positive correlation only between NE with IRS-1 (r = 0.72, p = 0.003), while in the OB-IR group, there was a positive correlation between the NE and A1AT with INSR (r = 0.62, p = 0.01 and r = 0.74, p = 0.002, respectively) and with IRS-2 (r = 0.74, p = 0.002 and r = 0.76, p = 0.001, respectively). CONCLUSION These results suggest that hyperinsulinemia and obesity are associated with changes in the expression of genes in leukocytes involved in the insulin pathway that are related to NE and A1AT.
Collapse
Affiliation(s)
- Walter David Cruz-Pineda
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Olga Lilia Garibay-Cerdenares
- CONACyT-Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Olga Lilia Garibay-Cerdenares, CONACyT-Universidad Autónoma de Guerrero, Avenida Lázaro Cárdenas S/N, Ciudad Universitaria, Chilpancingo, Guerrero, CP 39090, México, Tel/Fax +52 7474710901, Email
| | - Hugo Alberto Rodríguez-Ruíz
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Inés Matia-García
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Linda Anahí Marino-Ortega
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Mónica Espinoza-Rojo
- Laboratorio de Biología Molecular y Genómica, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Zyanya Reyes-Castillo
- Instituto de Investigaciones en Comportamiento Alimentario y Nutrición, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzmán, Jalisco, México
| | - Natividad Castro-Alarcón
- Laboratorio de Investigación en Microbiología, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Investigación en Biología Celular del Cáncer, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
| | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, México
- Correspondence: Isela Parra-Rojas, Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Avenida Lázaro Cárdenas S/N, Ciudad Universitaria, Chilpancingo, Guerrero, CP 39090, México, Tel/Fax +52 7474719310, Email
| |
Collapse
|
29
|
Cen HH, Hussein B, Botezelli JD, Wang S, Zhang JA, Noursadeghi N, Jessen N, Rodrigues B, Timmons JA, Johnson JD. Human and mouse muscle transcriptomic analyses identify insulin receptor mRNA downregulation in hyperinsulinemia-associated insulin resistance. FASEB J 2022; 36:e22088. [PMID: 34921686 PMCID: PMC9255858 DOI: 10.1096/fj.202100497rr] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 01/03/2023]
Abstract
Hyperinsulinemia is commonly viewed as a compensatory response to insulin resistance, yet studies have demonstrated that chronically elevated insulin may also drive insulin resistance. The molecular mechanisms underpinning this potentially cyclic process remain poorly defined, especially on a transcriptome-wide level. Transcriptomic meta-analysis in >450 human samples demonstrated that fasting insulin reliably and negatively correlated with INSR mRNA in skeletal muscle. To establish causality and study the direct effects of prolonged exposure to excess insulin in muscle cells, we incubated C2C12 myotubes with elevated insulin for 16 h, followed by 6 h of serum starvation, and established that acute AKT and ERK signaling were attenuated in this model of in vitro hyperinsulinemia. Global RNA-sequencing of cells both before and after nutrient withdrawal highlighted genes in the insulin receptor (INSR) signaling, FOXO signaling, and glucose metabolism pathways indicative of 'hyperinsulinemia' and 'starvation' programs. Consistently, we observed that hyperinsulinemia led to a substantial reduction in Insr gene expression, and subsequently a reduced surface INSR and total INSR protein, both in vitro and in vivo. Bioinformatic modeling combined with RNAi identified SIN3A as a negative regulator of Insr mRNA (and JUND, MAX, and MXI as positive regulators of Irs2 mRNA). Together, our analysis identifies mechanisms which may explain the cyclic processes underlying hyperinsulinemia-induced insulin resistance in muscle, a process directly relevant to the etiology and disease progression of type 2 diabetes.
Collapse
Affiliation(s)
- Haoning Howard Cen
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Bahira Hussein
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - José Diego Botezelli
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Su Wang
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jiashuo Aaron Zhang
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nilou Noursadeghi
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Brian Rodrigues
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - James A Timmons
- Augur Precision Medicine LTD, Stirling University Innovation Park, Stirling, Scotland.,William Harvey Research Institute, Queen Mary University of London, London, UK
| | - James D Johnson
- Department of Cellular and Physiological Sciences, Life Science Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Hsieh EJ, Lin WD, Schmidt W. Genomically Hardwired Regulation of Gene Activity Orchestrates Cellular Iron Homeostasis in Arabidopsis. RNA Biol 2021; 19:143-161. [PMID: 35067184 PMCID: PMC8786333 DOI: 10.1080/15476286.2021.2024024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/09/2021] [Accepted: 12/26/2021] [Indexed: 10/26/2022] Open
Abstract
Iron (Fe) is an essential micronutrient which plays pivotal roles as electron donor and catalyst across organisms. In plants, variable, often insufficient Fe supply necessitates mechanisms that constantly attune Fe uptake rates and recalibrate cellular Fe homoeostasis. Here, we show that short-term (0.5, 6, and 12 h) exposure of Arabidopsis thaliana plants to Fe deficiency triggered massive changes in gene activity governed by transcription and alternative splicing (AS), regulatory layers that were to a large extent mutually exclusive. Such preclusion was not observed for genes that are directly involved in the acquisition of Fe, which appears to be concordantly regulated by both expression and AS. Generally, genes with lower splice site strengths and higher intron numbers were more likely to be regulated by AS, no dependence on gene architecture was observed for transcriptionally controlled genes. Conspicuously, specific processes were associated with particular genomic features and biased towards either regulatory mode, suggesting that genomic hardwiring is functionally biased. Early changes in splicing patterns were, in many cases, congruent with later changes in transcript or protein abundance, thus contributing to the pronounced transcriptome-proteome discordance observed in plants.
Collapse
Affiliation(s)
- En-Jung Hsieh
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Wen-Dar Lin
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
| | - Wolfgang Schmidt
- Institute of Plant and Microbial Biology, Academia Sinica, Taipei, Taiwan
- Biotechnology Center, National Chung-Hsing University, Taichung, Taiwan
- Genome and Systems Biology Degree Program, College of Life Science, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
31
|
Sexual Dimorphism in Metabolic Responses to Western Diet in Drosophila melanogaster. Biomolecules 2021; 12:biom12010033. [PMID: 35053181 PMCID: PMC8774106 DOI: 10.3390/biom12010033] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022] Open
Abstract
Obesity is a chronic disease affecting millions of people worldwide. The fruit fly (Drosophila melanogaster) is an interesting research model to study metabolic and transcriptomic responses to obesogenic diets. However, the sex-specific differences in these responses are still understudied and perhaps underestimated. In this study, we exposed adult male and female Dahomey fruit flies to a standard diet supplemented with sugar, fat, or a combination of both. The exposure to a diet supplemented with 10% sugar and 10% fat efficiently induced an increase in the lipid content in flies, a hallmark for obesity. This increase in lipid content was more prominent in males, while females displayed significant changes in glycogen content. A strong effect of the diets on the ovarian size and number of ma-ture oocytes was also present in females exposed to diets supplemented with fat and a combina-tion of fat and sugar. In both males and females, fat body morphology changed and was associ-ated with an increase in lipid content of fat cells in response to the diets. The expression of me-tabolism-related genes also displayed a strong sexually dimorphic response under normal condi-tions and in response to sugar and/or fat-supplemented diets. Here, we show that the exposure of adult fruit flies to an obesogenic diet containing both sugar and fat allowed studying sexual dimorphism in metabolism and the expression of genes regulating metabolism.
Collapse
|
32
|
Nathanael J, Suardana P, Vianney YM, Dwi Putra SE. The role of FoxO1 and its modulation with small molecules in the development of diabetes mellitus: A review. Chem Biol Drug Des 2021; 99:344-361. [PMID: 34862852 DOI: 10.1111/cbdd.13989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/11/2021] [Accepted: 11/21/2021] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus type 2 (T2D) is one of the metabolic disorders suffered by a global human being. Certain factors, such as lifestyle and heredity, can increase a person's tendency for T2D. Various genes and proteins play a role in the development of insulin resistance and ultimately diabetes in which one central protein that is discussed in this review is FoxO1. In this review, we regard FoxO1 activation as detrimental, promote high plasma glucose level, and induce insulin resistance. Indeed, many contrasting studies arise since FoxO1 is an important protein to alleviate oxidative stress and promote cell survival, for example, also by preventing hyperglycemic-induced cell death. Inter-relation to PPARG, another important protein in metabolism, is also discussed. Ultimately, we discussed contrasting approaches of targeting FoxO1 to combat diabetes mellitus by small molecules.
Collapse
Affiliation(s)
- Joshua Nathanael
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Putu Suardana
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Yoanes Maria Vianney
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| | - Sulistyo Emantoko Dwi Putra
- Department of Biotechnology, Faculty of Biotechnology, University of Surabaya, Surabaya, East Java, Indonesia
| |
Collapse
|
33
|
Glial glucose fuels the neuronal pentose phosphate pathway for long-term memory. Cell Rep 2021; 36:109620. [PMID: 34433052 PMCID: PMC8411112 DOI: 10.1016/j.celrep.2021.109620] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 07/22/2021] [Accepted: 08/05/2021] [Indexed: 01/10/2023] Open
Abstract
Brain function relies almost solely on glucose as an energy substrate. The main model of brain metabolism proposes that glucose is taken up and converted into lactate by astrocytes to fuel the energy-demanding neuronal activity underlying plasticity and memory. Whether direct neuronal glucose uptake is required for memory formation remains elusive. We uncover, in Drosophila, a mechanism of glucose shuttling to neurons from cortex glia, an exclusively perisomatic glial subtype, upon formation of olfactory long-term memory (LTM). In vivo imaging reveals that, downstream of cholinergic activation of cortex glia, autocrine insulin signaling increases glucose concentration in glia. Glucose is then transferred from glia to the neuronal somata in the olfactory memory center to fuel the pentose phosphate pathway and allow LTM formation. In contrast, our results indicate that the increase in neuronal glucose metabolism, although crucial for LTM formation, is not routed to glycolysis. Neuronal glucose metabolism is increased upon long-term memory formation Glial cells shuttle glucose to neurons following insulin signaling activation Glucose fuels the neuronal pentose phosphate pathway
Collapse
|
34
|
Jahantigh D, Mirani Sargazi F, Sargazi S, Saravani R, Ghazaey Zidanloo S, Heidari Nia M, Piri M. Relationship between Functional miR-143/145 Cluster Variants and Susceptibility to Type 2 Diabetes Mellitus: A Preliminary Case-Control Study and Bioinformatics Analyses. Endocr Res 2021; 46:129-139. [PMID: 33870836 DOI: 10.1080/07435800.2021.1914079] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Purpose: To investigate the link between two variants (rs4705342 and rs4705343) in the promoter of the miR-143/145 cluster with Type 2 diabetes mellitus (T2DM) risk. Methods:A total of 1200 subjects were genotyped using the ARMS-PCR method. Results: The rs4705342 variant enhanced the risk of T2DM under codominant CC (OR = 3.24; 95% CI: 1.89-5.60), recessive TT+TC (OR = 3.02; 95% CI: 1.77-5.17), and dominant TC+CC (OR = 1.35; 95% CI: 1.08-1.71) genetic models. Individuals carrying the C allele of rs4705342 conferred a 1.43 fold increased risk of T2DM. As regards rs4705343, decreased risk of T2DM was observed under codominant TC (OR = 0.53; 95% CI: 0.42-0.67), over-dominant TT+CC (OR = 0.51; 95% CI: 0.40-0.64), and dominant TC+CC (OR = 0.59; 95% CI: 0.48-0.75) models. Haplotype analysis of the variants showed a 1.941-fold increased risk of T2DM regarding the C T combination. Significant associations were noticed between different haplotypes and lipid indices of T2DM patients. There were no notable changes in p-values after adjustment for BMI. Computational analysis revealed that miR143 and/or miR145 target important genes involved in glucose and lipid metabolism. Conclusions: Functional miR-143/145 variants might influence the risk of T2DM. Hence, clarifying the precise regulatory mechanisms of gene expression in the development of T2DM will significantly guide researchers to find a novel target for therapeutic intervention.
Collapse
Affiliation(s)
- Danial Jahantigh
- Department of Biology, Faculty of Science, University of Sistan and Baluchestan, Zahedan, Iran
| | - Fariba Mirani Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | | | - Milad Heidari Nia
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Maryam Piri
- Diabetes Center, Ali Asghar Hospital, Zahedan University of Medical Sciences, Zahedan, IR Iran
| |
Collapse
|
35
|
Roberto MP, Varano G, Vinas-Castells R, Holmes AB, Kumar R, Pasqualucci L, Farinha P, Scott DW, Dominguez-Sola D. Mutations in the transcription factor FOXO1 mimic positive selection signals to promote germinal center B cell expansion and lymphomagenesis. Immunity 2021; 54:1807-1824.e14. [PMID: 34380064 DOI: 10.1016/j.immuni.2021.07.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/26/2021] [Accepted: 07/13/2021] [Indexed: 12/27/2022]
Abstract
The transcription factor forkhead box O1 (FOXO1), which instructs the dark zone program to direct germinal center (GC) polarity, is typically inactivated by phosphatidylinositol 3-kinase (PI3K) signals. Here, we investigated how FOXO1 mutations targeting this regulatory axis in GC-derived B cell non-Hodgkin lymphomas (B-NHLs) contribute to lymphomagenesis. Examination of primary B-NHL tissues revealed that FOXO1 mutations and PI3K pathway activity were not directly correlated. Human B cell lines bearing FOXO1 mutations exhibited hyperactivation of PI3K and Stress-activated protein kinase (SAPK)/Jun amino-terminal kinase (JNK) signaling, and increased cell survival under stress conditions as a result of alterations in FOXO1 transcriptional affinities and activation of transcriptional programs characteristic of GC-positive selection. When modeled in mice, FOXO1 mutations conferred competitive advantage to B cells in response to key T-dependent immune signals, disrupting GC homeostasis. FOXO1 mutant transcriptional signatures were prevalent in human B-NHL and predicted poor clinical outcomes. Thus, rather than enforcing FOXO1 constitutive activity, FOXO1 mutations enable co-option of GC-positive selection programs during the pathogenesis of GC-derived lymphomas.
Collapse
Affiliation(s)
- Mark P Roberto
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gabriele Varano
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Rosa Vinas-Castells
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antony B Holmes
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Rahul Kumar
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY 10032, USA; Department of Pathology, Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032, USA
| | - Pedro Farinha
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC V5Z 1L3, Canada
| | - David W Scott
- Center for Lymphoid Cancer, British Columbia Cancer, Vancouver, BC V5Z 1L3, Canada
| | - David Dominguez-Sola
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
36
|
Shannon OM, Ashor AW, Scialo F, Saretzki G, Martin-Ruiz C, Lara J, Matu J, Griffiths A, Robinson N, Lillà L, Stevenson E, Stephan BCM, Minihane AM, Siervo M, Mathers JC. Mediterranean diet and the hallmarks of ageing. Eur J Clin Nutr 2021; 75:1176-1192. [PMID: 33514872 DOI: 10.1038/s41430-020-00841-x] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/09/2020] [Accepted: 12/07/2020] [Indexed: 12/21/2022]
Abstract
Ageing is a multifactorial process associated with reduced function and increased risk of morbidity and mortality. Recently, nine cellular and molecular hallmarks of ageing have been identified, which characterise the ageing process, and collectively, may be key determinants of the ageing trajectory. These include genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion and altered intercellular communication. Healthier dietary patterns reduce the risk of age-related diseases and increase longevity and may influence positively one or more of these hallmarks. The Mediterranean dietary pattern (MedDiet) is a plant-based eating pattern that was typical of countries such as Greece, Spain, and Italy pre-globalisation of the food system and which is associated with better health during ageing. Here we review the potential effects of a MedDiet on each of the nine hallmarks of ageing, and provide evidence that the MedDiet as a whole, or individual elements of this dietary pattern, may influence each hallmark positively-effects which may contribute to the beneficial effects of this dietary pattern on age-related disease risk and longevity. We also highlight potential avenues for future research.
Collapse
Affiliation(s)
- Oliver M Shannon
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne,, NE2 4HH, UK
| | - Ammar W Ashor
- Department of Pharmacology, College of Medicine, Al-Mustansiriyah University, Baghdad, Iraq
| | - Filippo Scialo
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne,, NE4 5PL, UK
- Dipartimento di Scienze Mediche Traslazionali, University of Campania "L. Vanvitelli", Naples, Italy
| | - Gabriele Saretzki
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne,, NE4 5PL, UK
| | - Carmen Martin-Ruiz
- Bioscience Institute, Bioscreening Core Facility, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne,, NE4 5PL, UK
| | - Jose Lara
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne,, NE1 8ST, UK
| | - Jamie Matu
- School of Clinical Applied Sciences, Leeds Beckett University, Leeds,, LS1 3HE, UK
| | - Alex Griffiths
- Institute for Sport, Physical Activity & Leisure, Leeds Beckett University, Leeds,, LS163QS, UK
| | - Natassia Robinson
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne,, NE4 5PL, UK
| | - Lionetti Lillà
- Department of Chemistry and Biology, University of Salerno, via Giovanni Paolo II, 132, 84084, Fisciano, SA, Italy
| | - Emma Stevenson
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne,, NE2 4HH, UK
| | - Blossom C M Stephan
- Institute of Mental Health, The University of Nottingham Medical School, Nottingham, UK
| | - Anne Marie Minihane
- Department of Nutrition and Preventive Medicine, Norwich Medical School, University of East Anglia (UEA), Norwich, UK
| | - Mario Siervo
- School of Life Sciences, The University of Nottingham Medical School, Queen's Medical Centre, Nottingham,, NG7 2UH, UK.
| | - John C Mathers
- Human Nutrition Research Centre, Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne,, NE2 4HH, UK
| |
Collapse
|
37
|
Al Baki MA, Kyo Jung J, Kim Y. Physiological Alterations in Deletion Mutants of Two Insulin-Like Peptides Encoded in Maruca vitrata Using CRISPR/Cas9. Front Physiol 2021; 12:701616. [PMID: 34276424 PMCID: PMC8284963 DOI: 10.3389/fphys.2021.701616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 06/09/2021] [Indexed: 11/15/2022] Open
Abstract
Most insect species encode multiple insulin-like peptides (ILPs) that exhibit functional overlaps in mediating physiological processes such as development and reproduction. Why do they need multiple ILPs? To address this question, we tested a hypothesis of the requirement of multiple ILPs by generating mutants lacking individual ILP genes using the CRISPR/Cas9 technology. Two ILPs (ILP1 and ILP2) in the legume pod borer, Maruca vitrata, mediate similar physiological processes such as hemolymph sugar level, larval development, and adult reproduction. Individual knock-out mutants (ΔILP1 and ΔILP2) were generated. They showed successful development from larvae to adults. However, they suffered from high hemolymph sugar levels by enhancing trehalose titers in the hemolymph. The hyperglycemic effect was more evident in ΔILP2 mutants than in ΔILP1 mutants. Both mutants showed increased expression of trehalose-6-phosphate synthase but suppressed expression of trehalase. These mutants also showed altered expression patterns of insulin signaling components. Expression levels of insulin receptor and Akt genes were upregulated, while those of FOXO and Target of rapamycin genes were downregulated in these mutants. These alterations of signal components resulted in significant retardation of immature development and reduced body sizes. ΔILP1 or ΔILP2 females exhibited poor oocyte development. Bromo-uridine incorporation was much reduced at the germarium of ovarioles of these mutants compared with wild females. Expression of the vitellogenin gene was also reduced in these mutants. Furthermore, males of these deletion mutants showed impaired reproductive activities when they mated with wild-type females. These results suggest that both ILPs are required for mediating larval development and adult reproduction in M. vitrata.
Collapse
Affiliation(s)
| | - Jin Kyo Jung
- Division of Crop Cultivation and Environment Research, Department of Central Area Crop Science, National Institute of Crop Science, Rural Development Administration, Suwon, South Korea
| | - Yonggyun Kim
- Department of Plant Medicals, Andong National University, Andong, South Korea
| |
Collapse
|
38
|
Ghomlaghi M, Hart A, Hoang N, Shin S, Nguyen LK. Feedback, Crosstalk and Competition: Ingredients for Emergent Non-Linear Behaviour in the PI3K/mTOR Signalling Network. Int J Mol Sci 2021; 22:6944. [PMID: 34203293 PMCID: PMC8267830 DOI: 10.3390/ijms22136944] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/20/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022] Open
Abstract
The PI3K/mTOR signalling pathway plays a central role in the governing of cell growth, survival and metabolism. As such, it must integrate and decode information from both external and internal sources to guide efficient decision-making by the cell. To facilitate this, the pathway has evolved an intricate web of complex regulatory mechanisms and elaborate crosstalk with neighbouring signalling pathways, making it a highly non-linear system. Here, we describe the mechanistic biological details that underpin these regulatory mechanisms, covering a multitude of negative and positive feedback loops, feed-forward loops, competing protein interactions, and crosstalk with major signalling pathways. Further, we highlight the non-linear and dynamic network behaviours that arise from these regulations, uncovered through computational and experimental studies. Given the pivotal role of the PI3K/mTOR network in cellular homeostasis and its frequent dysregulation in pathologies including cancer and diabetes, a coherent and systems-level understanding of the complex regulation and consequential dynamic signalling behaviours within this network is imperative for advancing biology and development of new therapeutic approaches.
Collapse
Affiliation(s)
- Milad Ghomlaghi
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Anthony Hart
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Nhan Hoang
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
| | - Sungyoung Shin
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Lan K. Nguyen
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences, Monash University, Clayton, VIC 3800, Australia; (M.G.); (A.H.); (N.H.); (S.S.)
- Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
39
|
McDonald JMC, Nabili P, Thorsen L, Jeon S, Shingleton AW. Sex-specific plasticity and the nutritional geometry of insulin-signaling gene expression in Drosophila melanogaster. EvoDevo 2021; 12:6. [PMID: 33990225 PMCID: PMC8120840 DOI: 10.1186/s13227-021-00175-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/17/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Sexual-size dimorphism (SSD) is replete among animals, but while the selective pressures that drive the evolution of SSD have been well studied, the developmental mechanisms upon which these pressures act are poorly understood. Ours and others' research has shown that SSD in D. melanogaster reflects elevated levels of nutritional plasticity in females versus males, such that SSD increases with dietary intake and body size, a phenomenon called sex-specific plasticity (SSP). Additional data indicate that while body size in both sexes responds to variation in protein level, only female body size is sensitive to variation in carbohydrate level. Here, we explore whether these difference in sensitivity at the morphological level are reflected by differences in how the insulin/IGF-signaling (IIS) and TOR-signaling pathways respond to changes in carbohydrates and proteins in females versus males, using a nutritional geometry approach. RESULTS The IIS-regulated transcripts of 4E-BP and InR most strongly correlated with body size in females and males, respectively, but neither responded to carbohydrate level and so could not explain the sex-specific response to body size to dietary carbohydrate. Transcripts regulated by TOR-signaling did, however, respond to dietary carbohydrate in a sex-specific manner. In females, expression of dILP5 positively correlated with body size, while expression of dILP2,3 and 8, was elevated on diets with a low concentration of both carbohydrate and protein. In contrast, we detected lower levels of dILP2 and 5 protein in the brains of females fed on low concentration diets. We could not detect any effect of diet on dILP expression in males. CONCLUSION Although females and males show sex-specific transcriptional responses to changes in protein and carbohydrate, the patterns of expression do not support a simple model of the regulation of body-size SSP by either insulin- or TOR-signaling. The data also indicate a complex relationship between carbohydrate and protein level, dILP expression and dILP peptide levels in the brain. In general, diet quality and sex both affect the transcriptional response to changes in diet quantity, and so should be considered in future studies that explore the effect of nutrition on body size.
Collapse
Affiliation(s)
- Jeanne M C McDonald
- Department of Ecology and Evolutionary Biology, Cornell University, Corson Hall Ithaca, NY, 14853, USA
- Department of Biology, Lake Forest College, 555 North Sheridan Road, Lake Forest, IL, 60045, USA
| | - Pegah Nabili
- Department of Biology, Lake Forest College, 555 North Sheridan Road, Lake Forest, IL, 60045, USA
| | - Lily Thorsen
- Department of Biology, Lake Forest College, 555 North Sheridan Road, Lake Forest, IL, 60045, USA
| | - Sohee Jeon
- Department of Biological Sciences, University of Illinois at Chicago, 840 W Taylor Street, Chicago, IL, 60607, USA
| | - Alexander W Shingleton
- Department of Biology, Lake Forest College, 555 North Sheridan Road, Lake Forest, IL, 60045, USA.
- Department of Biological Sciences, University of Illinois at Chicago, 840 W Taylor Street, Chicago, IL, 60607, USA.
| |
Collapse
|
40
|
Lee SM, Lee SW, Kang M, Choi JK, Park K, Byun JS, Kim DY. FoxO1 as a Regulator of Aquaporin 5 Expression in the Salivary Gland. J Dent Res 2021; 100:1281-1288. [PMID: 33840298 DOI: 10.1177/00220345211003490] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Forkhead box O1 (FoxO1) is a multifunctional initiator, mediator, and repressor of autoimmune diseases in an organ- or disease-specific manner. However, the role of FoxO1 in the salivary gland has not yet been elucidated. In this study, we discovered that FoxO1 and aquaporin 5 (AQP5) are both significantly downregulated in the patients with primary Sjögren syndrome, an autoimmune disease accompanying salivary gland dysfunction. Pharmacologic or genetic perturbation of FoxO1 in the rat salivary gland acinar cell line, SMG-C6, induced a significant downregulation of AQP5 expression, as observed in clinical specimens. There was a strong correlation between FoxO1 and AQP5 expression because FoxO1 is a direct regulator of AQP5 expression in salivary gland acinar cells through its interaction with the promoter region of AQP5. Serial injection of a FoxO1 inhibitor into mice induced a reduction of AQP5 expression in submandibular glands and, consequently, hyposalivation, which is one of the major clinical symptoms of primary Sjögren syndrome. However, there was no sign of inflammation or cell damage in the submandibular glands harvested from mice treated with the FoxO1 inhibitor. In conclusion, our findings indicate that FoxO1 in salivary gland tissue acts as a direct regulator of AQP5 expression. Thus, downregulation of FoxO1 observed in primary Sjögren syndrome is a putative mechanism for hyposalivation without the involvement of previously reported soluble factors in primary Sjögren syndrome patient sera.
Collapse
Affiliation(s)
- S M Lee
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - S W Lee
- Department of Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - M Kang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - J K Choi
- Department of Oral Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - K Park
- Department of Physiology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul, Republic of Korea
| | - J S Byun
- Department of Oral Medicine, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - D Y Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
41
|
Tandon S, Sarkar S. The S6k/4E-BP mediated growth promoting sub-pathway of insulin signalling cascade is essential to restrict pathogenesis of poly(Q) disorders in Drosophila. Life Sci 2021; 275:119358. [PMID: 33744321 DOI: 10.1016/j.lfs.2021.119358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/19/2021] [Accepted: 03/06/2021] [Indexed: 01/05/2023]
Abstract
Human neurodegenerative polyglutamine [poly(Q)] disorders, such as Huntington's disease (HD) and spinocerebellar ataxias (SCA), are characterised by an abnormal expansion of CAG repeats in the affected gene. The mutated proteins misfold and aggregate to form inclusion bodies that sequester important factors involved in cellular transcription, growth, stress and autophagic response and other essential functions. The insulin signalling pathway has been demonstrated as a major modifier and a potential drug target to ameliorate the poly(Q) mediated neurotoxicity in various model systems. Insulin signalling cascade harbours several downstream sub-pathways, which are synergistically involved in discharging indispensable biological functions such as growth and proliferation, metabolism, autophagy, regulation of cell death pathways etc. Hence, it is difficult to conclude whether the mitigation of poly(Q) neurotoxicity is an accumulative outcome of the insulin cascade, or the result of a specific sub-pathway. For the first time, we report that the ligand binding domain of insulin receptor mediated downstream growth promoting sub-pathway plays the pivotal role in operating the rescue event. We show that the growth promoting activity of insulin cascade is essential to minimize the abundance of inclusion bodies, to restrict neurodegeneration, and to restore the cellular transcriptional balance. Subsequently, we noted the involvement of the mTOR/S6k/4E-BP candidates in mitigating poly(Q) mediated neurotoxicity. Due to the conserved cellular functioning of the insulin cascade across species, and availability of several growth promoting molecules, our results in Drosophila poly(Q) models indicate towards a possibility of designing novel therapeutic strategies to restrict the pathogenesis of devastating human poly(Q) disorders.
Collapse
Affiliation(s)
- Shweta Tandon
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110 021, India
| | - Surajit Sarkar
- Department of Genetics, University of Delhi South Campus, Benito Juarez Road, New Delhi 110 021, India.
| |
Collapse
|
42
|
Rehman N, Varghese J. Larval nutrition influences adult fat stores and starvation resistance in Drosophila. PLoS One 2021; 16:e0247175. [PMID: 33606785 PMCID: PMC7895371 DOI: 10.1371/journal.pone.0247175] [Citation(s) in RCA: 16] [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: 08/07/2020] [Accepted: 02/02/2021] [Indexed: 12/16/2022] Open
Abstract
Insulin plays a major role in connecting nutrient availability to energy homeostasis by regulating metabolic pathways. Defects in insulin signalling is the primary cause for diabetes, obesity and various metabolic disorders. Nutritional status during growth and developmental stages play a crucial role in determining adult size, fecundity and ageing. However, the association between developmental nutrition and adult metabolic disorders has not been fully explored. Here, we address the effects of nutrient status during the larval growth phase on adult metabolism in Drosophila. We report that restricted food supply in larvae led to higher fat reserves and starvation resistance in mature adult flies, which we attribute to low insulin signalling. A lesser amount of stored fat was mobilised during early adult stages and during acute starvation, which accounts for the metabolic effects. Furthermore, larval diet influenced the expression of fat mobilisation genes brummer and lipid storage droplet-2 in adult flies, which led to the metabolic phenotypes reported here. Thus, the restricted nutrient environment in developing larvae led to adaptive changes that entrain the adult flies for scarce food availability.
Collapse
Affiliation(s)
- Niyas Rehman
- School of Biology, Indian Institute of Science Education and Research (IISER TVM), Thiruvananthapuram, Kerala, India
| | - Jishy Varghese
- School of Biology, Indian Institute of Science Education and Research (IISER TVM), Thiruvananthapuram, Kerala, India
- * E-mail:
| |
Collapse
|
43
|
Millington JW, Brownrigg GP, Chao C, Sun Z, Basner-Collins PJ, Wat LW, Hudry B, Miguel-Aliaga I, Rideout EJ. Female-biased upregulation of insulin pathway activity mediates the sex difference in Drosophila body size plasticity. eLife 2021; 10:e58341. [PMID: 33448263 PMCID: PMC7864645 DOI: 10.7554/elife.58341] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 01/11/2021] [Indexed: 12/14/2022] Open
Abstract
Nutrient-dependent body size plasticity differs between the sexes in most species, including mammals. Previous work in Drosophila showed that body size plasticity was higher in females, yet the mechanisms underlying increased female body size plasticity remain unclear. Here, we discover that a protein-rich diet augments body size in females and not males because of a female-biased increase in activity of the conserved insulin/insulin-like growth factor signaling pathway (IIS). This sex-biased upregulation of IIS activity was triggered by a diet-induced increase in stunted mRNA in females, and required Drosophila insulin-like peptide 2, illuminating new sex-specific roles for these genes. Importantly, we show that sex determination gene transformer promotes the diet-induced increase in stunted mRNA via transcriptional coactivator Spargel to regulate the male-female difference in body size plasticity. Together, these findings provide vital insight into conserved mechanisms underlying the sex difference in nutrient-dependent body size plasticity.
Collapse
Affiliation(s)
- Jason W Millington
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - George P Brownrigg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Charlotte Chao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Ziwei Sun
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Paige J Basner-Collins
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Lianna W Wat
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| | - Bruno Hudry
- MRC London Institute of Medical Sciences, and Institute of Clinical Sciences, Faculty of Medicine, Imperial College LondonLondonUnited Kingdom
| | - Irene Miguel-Aliaga
- MRC London Institute of Medical Sciences, and Institute of Clinical Sciences, Faculty of Medicine, Imperial College LondonLondonUnited Kingdom
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British ColumbiaVancouverCanada
| |
Collapse
|
44
|
Li YL, Yao YX, Zhao YM, Di YQ, Zhao XF. The steroid hormone 20-hydroxyecdysone counteracts insulin signaling via insulin receptor dephosphorylation. J Biol Chem 2021; 296:100318. [PMID: 33484713 PMCID: PMC7949120 DOI: 10.1016/j.jbc.2021.100318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 11/30/2022] Open
Abstract
The insulin receptor (INSR) binds insulin to promote body growth and maintain normal blood glucose levels. While it is known that steroid hormones such as estrogen and 20-hydroxyecdysone counteract insulin function, the molecular mechanisms responsible for this attenuation remain unclear. In the present study, using the agricultural pest lepidopteran Helicoverpa armigera as a model, we proposed that the steroid hormone 20-hydroxyecdysone (20E) induces dephosphorylation of INSR to counteract insulin function. We observed high expression and phosphorylation of INSR during larval feeding stages that decreased during metamorphosis. Insulin upregulated INSR expression and phosphorylation, whereas 20E repressed INSR expression and induced INSR dephosphorylation in vivo. Protein tyrosine phosphatase 1B (PTP1B, encoded by Ptpn1) dephosphorylated INSR in vivo. PTEN (phosphatase and tensin homolog deleted on chromosome 10) was critical for 20E-induced INSR dephosphorylation by maintaining the transcription factor Forkhead box O (FoxO) in the nucleus, where FoxO promoted Ptpn1 expression and repressed Insr expression. Knockdown of Ptpn1 using RNA interference maintained INSR phosphorylation, increased 20E production, and accelerated pupation. RNA interference of Insr in larvae repressed larval growth, decreased 20E production, delayed pupation, and accumulated hemolymph glucose levels. Taken together, these results suggest that a high 20E titer counteracts the insulin pathway by dephosphorylating INSR to stop larval growth and accumulate glucose in the hemolymph.
Collapse
Affiliation(s)
- Yan-Li Li
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - You-Xiang Yao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yu-Meng Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Yu-Qin Di
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiao-Fan Zhao
- Shandong Provincial Key Laboratory of Animal Cells and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China.
| |
Collapse
|
45
|
Sun CX, Li ZX. Biosynthesis of aphid alarm pheromone is modulated in response to starvation stress under regulation by the insulin, glycolysis and isoprenoid pathways. JOURNAL OF INSECT PHYSIOLOGY 2021; 128:104174. [PMID: 33242454 DOI: 10.1016/j.jinsphys.2020.104174] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 06/11/2023]
Abstract
The mechanism for biosynthesis and molecular regulation of the aphid alarm pheromone (AAP) is still a mystery. Previous studies indicated that the biosynthesis of AAP was directly affected by the terpenoid backbone biosynthesis pathway, and several pathways involved in nutritional metabolism providing the bricks for AAP biosynthesis were up-regulated in response to simulated stimulation. This suggests that AAP biosynthesis might be regulated by complex metabolic pathways. Here the molecular responses of the bird cherry-oat aphid Rhopalosiphum padi to starvation stress were investigated, and the molecular pathways were further analyzed by using RNA interference (RNAi) and protein inhibitor, combined with gas chromatography-mass spectrometry analysis of (E)-β-farnesene (EβF), the major component of the alarm pheromone in R. padi. The results showed that the nutritional stress significantly reduced the weight of aphid and the quantity of EβF, and meanwhile dramatically up-regulated the insulin receptor genes (InsR1/2) and down-regulated the downstream genes encoding the kinases PI3K and Akt, key enzymes in the glycolysis pathway (HK, A6PFK, PK) and the isoprenoid pathway (ACSS, HMGR, FPPS1, FPPS2, GGPPS, DPPS). PI3K inhibitor LY294002 treatment and RNAi-mediated knockdown of InsR1/2 significantly reduced the expression level of downstream genes and the quantity of EβF. Furthermore, knockdown of PK, the rate-limiting enzyme in the glycolysis pathway, down-regulated the genes in the isoprenoid pathway and the production of EβF; knockdown of the genes encoding isoprenyl diphosphate enzymes revealed that FPPS1 and FPPS2 were both required for EβF biosynthesis. Our data suggested that AAP is synthesized via glycolysis and isoprenoid pathways under regulation by the insulin signaling pathway.
Collapse
Affiliation(s)
- Cheng-Xian Sun
- Department of Entomology and MOA Key Laboratory for Monitoring and Environment-Friendly Control of Crop Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China
| | - Zheng-Xi Li
- Department of Entomology and MOA Key Laboratory for Monitoring and Environment-Friendly Control of Crop Pests, College of Plant Protection, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
46
|
Calissi G, Lam EWF, Link W. Therapeutic strategies targeting FOXO transcription factors. Nat Rev Drug Discov 2021; 20:21-38. [PMID: 33173189 DOI: 10.1038/s41573-020-0088-2] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 12/13/2022]
Abstract
FOXO proteins are transcription factors that are involved in numerous physiological processes and in various pathological conditions, including cardiovascular disease, cancer, diabetes and chronic neurological diseases. For example, FOXO proteins are context-dependent tumour suppressors that are frequently inactivated in human cancers, and FOXO3 is the second most replicated gene associated with extreme human longevity. Therefore, pharmacological manipulation of FOXO proteins is a promising approach to developing therapeutics for cancer and for healthy ageing. In this Review, we overview the role of FOXO proteins in health and disease and discuss the pharmacological approaches to modulate FOXO function.
Collapse
Affiliation(s)
- Giampaolo Calissi
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Imperial Centre for Translational and Experimental Medicine (ICTEM), London, UK
| | - Wolfgang Link
- Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), Madrid, Spain.
| |
Collapse
|
47
|
Nakamura M, Verboon JM, Allen TE, Abreu-Blanco MT, Liu R, Dominguez ANM, Delrow JJ, Parkhurst SM. Autocrine insulin pathway signaling regulates actin dynamics in cell wound repair. PLoS Genet 2020; 16:e1009186. [PMID: 33306674 PMCID: PMC7758051 DOI: 10.1371/journal.pgen.1009186] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 12/23/2020] [Accepted: 10/09/2020] [Indexed: 01/13/2023] Open
Abstract
Cells are exposed to frequent mechanical and/or chemical stressors that can compromise the integrity of the plasma membrane and underlying cortical cytoskeleton. The molecular mechanisms driving the immediate repair response launched to restore the cell cortex and circumvent cell death are largely unknown. Using microarrays and drug-inhibition studies to assess gene expression, we find that initiation of cell wound repair in the Drosophila model is dependent on translation, whereas transcription is required for subsequent steps. We identified 253 genes whose expression is up-regulated (80) or down-regulated (173) in response to laser wounding. A subset of these genes were validated using RNAi knockdowns and exhibit aberrant actomyosin ring assembly and/or actin remodeling defects. Strikingly, we find that the canonical insulin signaling pathway controls actin dynamics through the actin regulators Girdin and Chickadee (profilin), and its disruption leads to abnormal wound repair. Our results provide new insight for understanding how cell wound repair proceeds in healthy individuals and those with diseases involving wound healing deficiencies.
Collapse
Affiliation(s)
- Mitsutoshi Nakamura
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Jeffrey M. Verboon
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Tessa E. Allen
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Maria Teresa Abreu-Blanco
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Raymond Liu
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Andrew N. M. Dominguez
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Jeffrey J. Delrow
- Genomics Shared Resource, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Susan M. Parkhurst
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| |
Collapse
|
48
|
Zhang L, Zhang Z, Li C, Zhu T, Gao J, Zhou H, Zheng Y, Chang Q, Wang M, Wu J, Ran L, Wu Y, Miao H, Zou X, Liang B. S100A11 Promotes Liver Steatosis via FOXO1-Mediated Autophagy and Lipogenesis. Cell Mol Gastroenterol Hepatol 2020; 11:697-724. [PMID: 33075563 PMCID: PMC7841444 DOI: 10.1016/j.jcmgh.2020.10.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Nonalcoholic fatty liver disease (NAFLD) is becoming a severe liver disorder worldwide. Autophagy plays a critical role in liver steatosis. However, the role of autophagy in NAFLD remains exclusive and under debate. In this study, we investigated the role of S100 calcium binding protein A11 (S100A11) in the pathogenesis of hepatic steatosis. METHODS We performed liver proteomics in a well-established tree shrew model of NAFLD. The expression of S100A11 in different models of NAFLD was detected by Western blot and/or quantitative polymerase chain reaction. Liver S100A11 overexpression mice were generated by injecting a recombinant adenovirus gene transfer vector through the tail vein and then induced by a high-fat and high-cholesterol diet. Cell lines with S100a11 stable overexpression were established with a recombinant lentiviral vector. The lipid content was measured with either Bodipy staining, Oil Red O staining, gas chromatography, or a triglyceride kit. The autophagy and lipogenesis were detected in vitro and in vivo by Western blot and quantitative polymerase chain reaction. The functions of Sirtuin 1, histone deacetylase 6 (HDAC6), and FOXO1 were inhibited by specific inhibitors. The interactions between related proteins were analyzed by a co-immunoprecipitation assay and immunofluorescence analysis. RESULTS The expression of S100A11 was up-regulated significantly in a time-dependent manner in the tree shrew model of NAFLD. S100A11 expression was induced consistently in oleic acid-treated liver cells as well as the livers of mice fed a high-fat diet and NAFLD patients. Both in vitro and in vivo overexpression of S100A11 could induce hepatic lipid accumulation. Mechanistically, overexpression of S100A11 activated an autophagy and lipogenesis process through up-regulation and acetylation of the transcriptional factor FOXO1, consequently promoting lipogenesis and lipid accumulation in vitro and in vivo. Inhibition of HDAC6, a deacetylase of FOXO1, showed similar phenotypes to S100A11 overexpression in Hepa 1-6 cells. S100A11 interacted with HDAC6 to inhibit its activity, leading to the release and activation of FOXO1. Under S100A11 overexpression, the inhibition of FOXO1 and autophagy could alleviate the activated autophagy as well as up-regulated lipogenic genes. Both FOXO1 and autophagy inhibition and Dgat2 deletion could reduce liver cell lipid accumulation significantly. CONCLUSIONS A high-fat diet promotes liver S100A11 expression, which may interact with HDAC6 to block its binding to FOXO1, releasing or increasing the acetylation of FOXO1, thus activating autophagy and lipogenesis, and accelerating lipid accumulation and liver steatosis. These findings indicate a completely novel S100A11-HDAC6-FOXO1 axis in the regulation of autophagy and liver steatosis, providing potential possibilities for the treatment of NAFLD.
Collapse
Affiliation(s)
- Linqiang Zhang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Zhiguo Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Chengbin Li
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China
| | - Tingting Zhu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Jing Gao
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hu Zhou
- Department of Analytical Chemistry and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Yingzhuan Zheng
- College of Life Sciences, Yunnan Normal University, Kunming, Yunnan, China
| | - Qing Chang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China; Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Mingshan Wang
- Howard Hughes Medical Institute, Department of Ecology and Evolutionary Biology, University of California Santa Cruz, Santa Cruz, California
| | - Jieyu Wu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Liyuan Ran
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China
| | - Yingjie Wu
- Institute for Genome Engineered Animal Models of Human Diseases, Dalian Medical University, Dalian, Liaoning, China; Shandong Laboratory Animal Center, Science and Technology Innovation Center, Shandong Provincial Hospital, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Huilai Miao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Xiaoju Zou
- School of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China.
| | - Bin Liang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan, China; Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China; Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China.
| |
Collapse
|
49
|
Texada MJ, Koyama T, Rewitz K. Regulation of Body Size and Growth Control. Genetics 2020; 216:269-313. [PMID: 33023929 PMCID: PMC7536854 DOI: 10.1534/genetics.120.303095] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 06/29/2020] [Indexed: 12/20/2022] Open
Abstract
The control of body and organ growth is essential for the development of adults with proper size and proportions, which is important for survival and reproduction. In animals, adult body size is determined by the rate and duration of juvenile growth, which are influenced by the environment. In nutrient-scarce environments in which more time is needed for growth, the juvenile growth period can be extended by delaying maturation, whereas juvenile development is rapidly completed in nutrient-rich conditions. This flexibility requires the integration of environmental cues with developmental signals that govern internal checkpoints to ensure that maturation does not begin until sufficient tissue growth has occurred to reach a proper adult size. The Target of Rapamycin (TOR) pathway is the primary cell-autonomous nutrient sensor, while circulating hormones such as steroids and insulin-like growth factors are the main systemic regulators of growth and maturation in animals. We discuss recent findings in Drosophila melanogaster showing that cell-autonomous environment and growth-sensing mechanisms, involving TOR and other growth-regulatory pathways, that converge on insulin and steroid relay centers are responsible for adjusting systemic growth, and development, in response to external and internal conditions. In addition to this, proper organ growth is also monitored and coordinated with whole-body growth and the timing of maturation through modulation of steroid signaling. This coordination involves interorgan communication mediated by Drosophila insulin-like peptide 8 in response to tissue growth status. Together, these multiple nutritional and developmental cues feed into neuroendocrine hubs controlling insulin and steroid signaling, serving as checkpoints at which developmental progression toward maturation can be delayed. This review focuses on these mechanisms by which external and internal conditions can modulate developmental growth and ensure proper adult body size, and highlights the conserved architecture of this system, which has made Drosophila a prime model for understanding the coordination of growth and maturation in animals.
Collapse
Affiliation(s)
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, 2100, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, 2100, Denmark
| |
Collapse
|
50
|
Dos Santos MM, de Macedo GT, Prestes AS, Ecker A, Müller TE, Leitemperger J, Fontana BD, Ardisson-Araújo DMP, Rosemberg DB, Barbosa NV. Modulation of redox and insulin signaling underlie the anti-hyperglycemic and antioxidant effects of diphenyl diselenide in zebrafish. Free Radic Biol Med 2020; 158:20-31. [PMID: 32544425 DOI: 10.1016/j.freeradbiomed.2020.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/01/2020] [Accepted: 06/01/2020] [Indexed: 12/12/2022]
Abstract
The organic selenium compound diphenyl diselenide (DD) has been recognized as an antioxidant and neuroprotective agent, exerting an anti-hyperglycemic effect in experimental models of diabetes. However, the precise mechanisms involved in the protection are unclear. Using the zebrafish (Danio rerio) as a model organism, here we investigated biomarkers underlying the protective effects of DD against hyperglycemia, targeting in a transcriptional approach the redox and insulin-signaling pathway. Fish were fed on a diet containing DD (3 mg/kg) for 74 days. In the last 14 days, they were exposed to a 111 mM glucose solution to induce a hyperglycemic state. DD reduced blood glucose levels as well as normalized the brain mRNA transcription of four insulin receptors-coding genes (Insra1, Insra2, Insrb1, Insrb2), which were down-regulated by glucose. DD alone caused an up-regulation of relative mRNA transcription in both Insra receptors and glucose transporter 3 genes. DD counteracted hyperglycemia-induced lipid peroxidation, protein and thiol depletion. Along with the decreased activity of antioxidant enzymes SOD and GPx, the brain of hyperglycemic fish presented a reduction in mRNA transcription of FoxO3A, FoxO3B, Nrf2, GPx3A, SOD1, and SOD2 genes. Besides normalizing the transcriptional levels, DD caused an up-regulation of relative mRNAs that encode Nrf2, FoxO1A, FOXO3A, GPx4A, PTP1B, AKT and SelP. Collectively, our findings suggest that the antioxidant and anti-hyperglycemic actions of DD in a zebrafish diabetes model are likely associated with the regulation of the oxidative stress resistance and the insulin-signaling pathway and that could be related to the modulation at mRNA level of two important transcription factors, Nrf2 and FoxO.
Collapse
Affiliation(s)
- Matheus M Dos Santos
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Gabriel T de Macedo
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Alessandro S Prestes
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Assis Ecker
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Talise E Müller
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Jossiele Leitemperger
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Bárbara D Fontana
- Brain and Behaviour Laboratory, School of Pharmacy and Biomedical Sciences, University of Portsmouth, England, UK
| | - Daniel M P Ardisson-Araújo
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil
| | - Denis B Rosemberg
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil; Zebrafish Neuroscience Research Consortium (ZNRC), 309 Palmer Court, Slidell, LA, 70458, USA
| | - Nilda V Barbosa
- Programa de Pós-graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, 1000, 97105-900, Santa Maria, RS, Brazil.
| |
Collapse
|