1
|
Frappaolo A, Giansanti MG. Using Drosophila melanogaster to Dissect the Roles of the mTOR Signaling Pathway in Cell Growth. Cells 2023; 12:2622. [PMID: 37998357 PMCID: PMC10670727 DOI: 10.3390/cells12222622] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/10/2023] [Accepted: 11/11/2023] [Indexed: 11/25/2023] Open
Abstract
The evolutionarily conserved target of rapamycin (TOR) serine/threonine kinase controls eukaryotic cell growth, metabolism and survival by integrating signals from the nutritional status and growth factors. TOR is the catalytic subunit of two distinct functional multiprotein complexes termed mTORC1 (mechanistic target of rapamycin complex 1) and mTORC2, which phosphorylate a different set of substrates and display different physiological functions. Dysregulation of TOR signaling has been involved in the development and progression of several disease states including cancer and diabetes. Here, we highlight how genetic and biochemical studies in the model system Drosophila melanogaster have been crucial to identify the mTORC1 and mTORC2 signaling components and to dissect their function in cellular growth, in strict coordination with insulin signaling. In addition, we review new findings that involve Drosophila Golgi phosphoprotein 3 in regulating organ growth via Rheb-mediated activation of mTORC1 in line with an emerging role for the Golgi as a major hub for mTORC1 signaling.
Collapse
Affiliation(s)
- Anna Frappaolo
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| | - Maria Grazia Giansanti
- Istituto di Biologia e Patologia Molecolari del CNR, c/o Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, 00185 Roma, Italy
| |
Collapse
|
2
|
Zhou Y, Zhou L, Li Q, Zhu X, Yu Z, Ke H, Chen Q, Ren B. Transcriptome analysis and identification of genes related to environmental adaptation of Grylloprimevala jilina Zhou & Ren 2023. Ecol Evol 2023; 13:e10717. [PMID: 38020696 PMCID: PMC10659822 DOI: 10.1002/ece3.10717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 10/08/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Grylloprimevala jilina is a true cave insect living in the dark areas of caves. It has the characteristics of sparse skin pigmentation, degeneration of the compound eyes and monocular eyes, and obvious preference for high-humidity and low-temperature environments. Given the highly specialized, rare, and limited distribution, G. jilina is considered an endangered species and also a first-level national protected insect in China. Cave creatures often undergo dramatic morphological changes in their sensory systems to adapt to the cave environment. Most previous studies mainly focused on morphological adaptive changes in cave insects, and only a few studied the changes at the gene level. In this study, we performed transcriptome analysis of G. jilina and constructed phylogenetic trees of genes that are related to environmental adaptation, including chemosensory, visual-related, reproduction-related, temperature adaptation-related, and winged morph differentiation-related genes. Besides, the expression levels of environmental adaption-related genes in different tissues, including antennae, heads, thoraxes, abdomens, legs, and tails, were analyzed. The results showed the loss of chemosensory genes and vision-related genes, the conservation of reproduction-related genes and temperature adaptation-related genes, and the conservation of wing-related genes despite the loss of wings, and the results were consistent with other cave insects. The identification and expression study of genes possibly related to the environmental adaptability in G. jilina provided basic data for the protection of this endangered species and increased knowledge about insect evolution in general.
Collapse
Affiliation(s)
- Yuxin Zhou
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
| | - Lin Zhou
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
- Istitude of Plant Protection Jilim Academy of Agricultural Science/Jilin Key Laboratory of Agricultural Microbiology/Key Laboratory of Integrated Pest Management on Crops in Northeast ChinaMinistry of Agriculture and Rural AreasGongzhlingChina
| | - Qiuyao Li
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
| | - Xiaoyan Zhu
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
| | - Zhongbo Yu
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
| | - Haoqin Ke
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
| | - Qi Chen
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
| | - Bingzhong Ren
- Jilin Provincial Key Laboratory of Animal Resource Conservation and UtilizationNortheast Normal UniversityChangchunChina
- Key Laboratory of Vegetation Ecology, MOENortheast Normal UniversityChangchunChina
| |
Collapse
|
3
|
Abstract
Nutrient intake is obligatory for animal growth and development, but nutrients alone are not sufficient. Indeed, insulin and homologous hormones are required for normal growth even in the presence of nutrients. These hormones communicate nutrient status between organs, allowing animals to coordinate growth and metabolism with nutrient supply. Insulin and related hormones, such as insulin-like growth factors and insulin-like peptides, play important roles in development and metabolism, with defects in insulin production and signaling leading to hyperglycemia and diabetes. Here, we describe the insulin hormone family and the signal transduction pathways activated by these hormones. We highlight the roles of insulin signaling in coordinating maternal and fetal metabolism and growth during pregnancy, and we describe how secretion of insulin is regulated at different life stages. Additionally, we discuss the roles of insulin signaling in cell growth, stem cell proliferation and cell differentiation. We provide examples of the role of insulin in development across multiple model organisms: Caenorhabditis elegans, Drosophila, zebrafish, mouse and human.
Collapse
Affiliation(s)
- Miyuki Suzawa
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| | - Michelle L. Bland
- Department of Pharmacology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
4
|
Xing BL, Wang SF, Gulinuer A, Ye GY, Yang L. Transcriptional regulation of host insulin signaling pathway genes controlling larval development by Microplitis manilae parasitization. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2023; 113:e22003. [PMID: 36694471 DOI: 10.1002/arch.22003] [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: 12/17/2022] [Revised: 01/09/2023] [Accepted: 01/15/2023] [Indexed: 06/17/2023]
Abstract
Idiobiont parasitoids using other insects as hosts sabotage the host growth and development to ensure their offspring survival. Numerous studies have discovered that insect development is subtly regulated by the conserved insulin signaling pathway. However, little is known about how wasp parasitization disrupts host development controlled by the insulin signaling pathway. Here we address this study to determine the effect of wasp parasitism on host Spodoptera frugiperda development using the idiobiont parasitoid Microplitis manilae as a model. Upon M. manilae parasitization, the body weight, body length, and food consumption of host insect were dramatically reduced compared to the unparasitized S. frugiperda. We next identified the core genes involved in host insulin signaling pathway and further analyzed the domain organizations of these genes. Phylogenetic reconstruction based on the insulin receptors clustered S. frugiperda together with other noctuidae insects. In the latter study, we profiled the expression patterns of host insulin signaling pathway genes in response to M. manilae parasitization at 2, 24, and 48 h, significant decreases in mRNA levels were recorded in S. frugiperda larvae upon 24 and 48 h parasitization. These current findings substantially add to our understanding of the physiological interaction between parasitoid and host insects, thus contributing to revealing the molecular mechanism of parasitic wasps regulating host development.
Collapse
Affiliation(s)
- Bing-Lin Xing
- Hainan Institute, Zhejiang University, Sanya, China
- Sanya Nanfan Research Institute & School of Tropical Crops, Hainan University, Sanya, China
| | - Shao-Feng Wang
- Hainan Institute, Zhejiang University, Sanya, China
- State Key Laboratory of Rice Biology & Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Ahamaijiang Gulinuer
- Sanya Nanfan Research Institute & School of Tropical Crops, Hainan University, Sanya, China
| | - Gong-Yin Ye
- Hainan Institute, Zhejiang University, Sanya, China
- State Key Laboratory of Rice Biology & Ministry of Agriculture Key Lab of Molecular Biology of Crop Pathogens and Insects, Institute of Insect Sciences, Zhejiang University, Hangzhou, China
| | - Lei Yang
- Sanya Nanfan Research Institute & School of Tropical Crops, Hainan University, Sanya, China
| |
Collapse
|
5
|
Yan L, Du H, Li Y, Li X, Sun L, Cao C. Identification and characterization of key genes in insulin signaling pathway as molecular targets for controlling the fall webworm, Hyphantria cunea. PEST MANAGEMENT SCIENCE 2023; 79:899-908. [PMID: 36317953 DOI: 10.1002/ps.7268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 10/22/2022] [Accepted: 10/28/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND The insulin signaling pathway is closely related to metabolism, growth, reproductive capacity and lifespan of insects. However, the physiological function of the insulin signaling pathway is little known in Hyphantria cunea. RESULTS Five insulin signaling pathway genes (HcInR, HcPI3K, HcAKT, HcFOXO and HcTOR) in H. cunea were identified and characterized in this study. The spatiotemporal expression profiles of the genes showed that HcInR, HcAKT, HcPI3K and HcTOR expressions were higher at the egg stage than those in other development stages, whereas HcFOXO was highly expressed in the adult stage; all of these genes were highly expressed in the larval digestive system, especially in the midgut and hindgut. After RNA interference (RNAi) of the five genes in 5th instar H. cunea larvae, weight gain and survival rate (except in the siHcAKT-injected group) were significantly decreased, and the developmental duration of larval and pupal stages were prolonged. In addition, knockdown of five genes in 7th instar larvae decreased the pupation rate, survival rate and oviposition capacity, and resulted in abnormal development during larval-pupal transition. CONCLUSION Our findings indicate that the insulin signaling pathway plays essential roles in growth and development and the molting process in H. cunea, providing an important basis for developing new potentially molecular targets for RNAi-based pest control and understanding the mechanism of H. cunea outbreak. © 2022 Society of Chemical Industry.
Collapse
Affiliation(s)
- Liqiong Yan
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin, China
| | - Hui Du
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin, China
| | - Ye Li
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin, China
| | - Xue Li
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin, China
| | - Lili Sun
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin, China
| | - Chuanwang Cao
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin, China
| |
Collapse
|
6
|
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: 6.0] [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
|
7
|
Xu P, Zheng Y, Liao J, Hu M, Yang Y, Zhang B, Kilby MD, Fu H, Liu Y, Zhang F, Xiong L, Liu X, Jin H, Wu Y, Huang J, Han T, Wen L, Gao R, Fu Y, Fan X, Qi H, Baker PN, Tong C. AMPK regulates homeostasis of invasion and viability in trophoblasts by redirecting glucose metabolism: Implications for pre-eclampsia. Cell Prolif 2022; 56:e13358. [PMID: 36480593 PMCID: PMC9890534 DOI: 10.1111/cpr.13358] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 12/13/2022] Open
Abstract
Pre-eclampsia (PE) is deemed an ischemia-induced metabolic disorder of the placenta due to defective invasion of trophoblasts during placentation; thus, the driving role of metabolism in PE pathogenesis is largely ignored. Since trophoblasts undergo substantial glycolysis, this study aimed to investigate its function and regulatory mechanism by AMPK in PE development. Metabolomics analysis of PE placentas was performed by gas chromatography-mass spectrometry (GC-MS). Trophoblast-specific AMPKα1-deficient mouse placentas were generated to assess morphology. A mouse PE model was established by Reduced Uterine Perfusion Pressure, and placental AMPK was modulated by nanoparticle-delivered A769662. Trophoblast glucose uptake was measured by 2-NBDG and 2-deoxy-d-[3 H] glucose uptake assays. Cellular metabolism was investigated by the Seahorse assay and GC-MS.PE complicated trophoblasts are associated with AMPK hyperactivation due not to energy deficiency. Thereafter, AMPK activation during placentation exacerbated PE manifestations but alleviated cell death in the placenta. AMPK activation in trophoblasts contributed to GLUT3 translocation and subsequent glucose metabolism, which were redirected into gluconeogenesis, resulting in deposition of glycogen and accumulation of phosphoenolpyruvate; the latter enhanced viability but compromised trophoblast invasion. However, ablation of AMPK in the mouse placenta resulted in decreased glycogen deposition and structural malformation. These data reveal a novel homeostasis between invasiveness and viability in trophoblasts, which is mechanistically relevant for switching between the 'go' and 'grow' cellular programs.
Collapse
Affiliation(s)
- Ping Xu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina,Biochemistry and Molecular BiologyUniversity of Texas McGovern Medical SchoolHoustonTexasUSA
| | - Yangxi Zheng
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina,Department of Stem Cell Transplantation and Cell TherapyMD Anderson Cancer CenterHoustonTexasUSA
| | - Jiujiang Liao
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Mingyu Hu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Yike Yang
- Department of Gynecology and ObstetricsPeking University Third HospitalBeijingChina
| | - Baozhen Zhang
- Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Mark D. Kilby
- Institute of Metabolism and System ResearchUniversity of BirminghamEdgbastonUK
| | - Huijia Fu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Yamin Liu
- Department of ObstetricsWomen and Children's Hospital of Chongqing Medical UniversityChongqingChina
| | - Fumei Zhang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Liling Xiong
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Xiyao Liu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Huili Jin
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Yue Wu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Jiayu Huang
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Tingli Han
- Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Li Wen
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Rufei Gao
- Laboratory of Reproductive Biology, School of Public Health and ManagementChongqing Medical UniversityChongqingChina
| | - Yong Fu
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| | - Xiujun Fan
- Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Hongbo Qi
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina,Department of ObstetricsWomen and Children's Hospital of Chongqing Medical UniversityChongqingChina
| | | | - Chao Tong
- State Key Laboratory of Maternal and Fetal Medicine of Chongqing MunicipalityThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina,Ministry of Education‐International Collaborative Laboratory of Reproduction and DevelopmentChongqing Medical UniversityChongqingChina
| |
Collapse
|
8
|
Haroon, Li YX, Ye CX, Su J, Nabi G, Su XH, Xing LX. De Novo Transcriptome Assembly and Analysis of Longevity Genes Using Subterranean Termite ( Reticulitermes chinensis) Castes. Int J Mol Sci 2022; 23:13660. [PMID: 36362447 PMCID: PMC9657995 DOI: 10.3390/ijms232113660] [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: 08/05/2022] [Revised: 10/20/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022] Open
Abstract
The longevity phenomenon is entirely controlled by the insulin signaling pathway (IIS-pathway). Both vertebrates and invertebrates have IIS-pathways that are comparable to one another, though no one has previously described de novo transcriptome assembly of IIS-pathway-associated genes in termites. In this research, we analyzed the transcriptomes of both reproductive (primary kings “PK” and queens “PQ”, secondary worker reproductive kings “SWRK” and queens “SWRQ”) and non-reproductive (male “WM” and female “WF” workers) castes of the subterranean termite Reticulitermes chinensis. The goal was to identify the genes responsible for longevity in the reproductive and non-reproductive castes. Through transcriptome analysis, we annotated 103,589,264 sequence reads and 184,436 (7G) unigenes were assembled, GC performance was measured at 43.02%, and 64,046 sequences were reported as CDs sequences. Of which 35 IIS-pathway-associated genes were identified, among 35 genes, we focused on the phosphoinositide-dependent kinase-1 (Pdk1), protein kinase B2 (akt2-a), tuberous sclerosis-2 (Tsc2), mammalian target of rapamycin (mTOR), eukaryotic translation initiation factor 4E (EIF4E) and ribosomal protein S6 (RPS6) genes. Previously these genes (Pdk1, akt2-a, mTOR, EIF4E, and RPS6) were investigated in various organisms, that regulate physiological effects, growth factors, protein translation, cell survival, proliferation, protein synthesis, cell metabolism and survival, autophagy, fecundity rate, egg size, and follicle number, although the critical reason for longevity is still unclear in the termite castes. However, based on transcriptome profiling, the IIS-pathway-associated genes could prolong the reproductive caste lifespan and health span. Therefore, the transcriptomic shreds of evidence related to IIS-pathway genes provide new insights into the maintenance and relationships between biomolecular homeostasis and remarkable longevity. Finally, we propose a strategy for future research to decrypt the hidden costs associated with termite aging in reproductive and non-reproductive castes.
Collapse
Affiliation(s)
- Haroon
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Yu-Xin Li
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Chen-Xu Ye
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Jian Su
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
| | - Ghulam Nabi
- Institute of Nature Conservation, Polish Academy of Sciences, 31120 Krakow, Poland
| | - Xiao-Hong Su
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China
| | - Lian-Xi Xing
- College of Life Sciences, Northwest University, No. 229, North Taibai Rd., Xi’an 710069, China
- Shaanxi Key Laboratory for Animal Conservation, Northwest University, Xi’an 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Northwest University, Xi’an 710069, China
| |
Collapse
|
9
|
Ogienko AA, Omelina ES, Bylino OV, Batin MA, Georgiev PG, Pindyurin AV. Drosophila as a Model Organism to Study Basic Mechanisms of Longevity. Int J Mol Sci 2022; 23:11244. [PMID: 36232546 PMCID: PMC9569508 DOI: 10.3390/ijms231911244] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
The spatio-temporal regulation of gene expression determines the fate and function of various cells and tissues and, as a consequence, the correct development and functioning of complex organisms. Certain mechanisms of gene activity regulation provide adequate cell responses to changes in environmental factors. Aside from gene expression disorders that lead to various pathologies, alterations of expression of particular genes were shown to significantly decrease or increase the lifespan in a wide range of organisms from yeast to human. Drosophila fruit fly is an ideal model system to explore mechanisms of longevity and aging due to low cost, easy handling and maintenance, large number of progeny per adult, short life cycle and lifespan, relatively low number of paralogous genes, high evolutionary conservation of epigenetic mechanisms and signalling pathways, and availability of a wide range of tools to modulate gene expression in vivo. Here, we focus on the organization of the evolutionarily conserved signaling pathways whose components significantly influence the aging process and on the interconnections of these pathways with gene expression regulation.
Collapse
Affiliation(s)
- Anna A. Ogienko
- Department of Regulation of Genetic Processes, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia
| | - Evgeniya S. Omelina
- Department of Regulation of Genetic Processes, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia
- Laboratory of Biotechnology, Novosibirsk State Agrarian University, 630039 Novosibirsk, Russia
| | - Oleg V. Bylino
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology RAS, 119334 Moscow, Russia
| | - Mikhail A. Batin
- Open Longevity, 15260 Ventura Blvd., Sherman Oaks, Los Angeles, CA 91403, USA
| | - Pavel G. Georgiev
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology RAS, 119334 Moscow, Russia
| | - Alexey V. Pindyurin
- Department of Regulation of Genetic Processes, Institute of Molecular and Cellular Biology SB RAS, 630090 Novosibirsk, Russia
| |
Collapse
|
10
|
Fumagalli S, Pende M. S6 kinase 1 at the central node of cell size and ageing. Front Cell Dev Biol 2022; 10:949196. [PMID: 36036012 PMCID: PMC9417411 DOI: 10.3389/fcell.2022.949196] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Genetic evidence in living organisms from yeast to plants and animals, including humans, unquestionably identifies the Target Of Rapamycin kinase (TOR or mTOR for mammalian/mechanistic) signal transduction pathway as a master regulator of growth through the control of cell size and cell number. Among the mTOR targets, the activation of p70 S6 kinase 1 (S6K1) is exquisitely sensitive to nutrient availability and rapamycin inhibition. Of note, in vivo analysis of mutant flies and mice reveals that S6K1 predominantly regulates cell size versus cell proliferation. Here we review the putative mechanisms of S6K1 action on cell size by considering the main functional categories of S6K1 targets: substrates involved in nucleic acid and protein synthesis, fat mass accumulation, retrograde control of insulin action, senescence program and cytoskeleton organization. We discuss how S6K1 may be involved in the observed interconnection between cell size, regenerative and ageing responses.
Collapse
Affiliation(s)
| | - Mario Pende
- *Correspondence: Stefano Fumagalli, ; Mario Pende,
| |
Collapse
|
11
|
Morgan A, Kiser CA, Bronson I, Lin H, Guillette N, McMahon R, Kennell JA, Long LJ, Reed LK, Rele CP. Drosophila eugracilis - Akt. MICROPUBLICATION BIOLOGY 2022; 2022:10.17912/micropub.biology.000544. [PMID: 35856017 PMCID: PMC9287739 DOI: 10.17912/micropub.biology.000544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/06/2022]
Abstract
Gene Model for Akt in the D. eugracilis (DeugGB2) assembly (GCA_000236325.2).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Chinmay P. Rele
- The University of Alabama, Tuscaloosa, AL USA
,
Correspondence to: Chinmay P. Rele (
)
| |
Collapse
|
12
|
Kim YJ. Activity-induced synaptic structural modifications by Akt. Biochem Biophys Res Commun 2022; 621:94-100. [PMID: 35820284 DOI: 10.1016/j.bbrc.2022.06.093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022]
Abstract
The activity-dependent regulation of synaptic structures plays a key role in synaptic development and plasticity; however, the signaling mechanisms involved remain largely unknown. The serine/threonine protein kinase Akt, a downstream effector of phosphoinositide 3-kinase (PI3K), plays a pivotal role in a wide range of physiological functions. We focused on the importance of Akt in rapid synaptic structural changes after stimulation at the Drosophila neuromuscular junction, a well-studied model synapse. Compared with wild-type larvae, akt mutants showed significantly reduced muscle size and an increased number of boutons per area, suggesting that Akt is required for proper pre- and postsynaptic growth. In addition, the level of cysteine string protein (CSP) was significantly increased, and its distribution was different in akt mutants. After high K+ single stimulation, the CSP level of akt mutant NMJs increased dramatically compared with that of wild-type NMJs. Interestingly, ghost boutons without postsynaptic specialization were found in akt mutant NMJs, and the number of these boutons was significantly increased by patterned stimulation. In contrast, the postsynaptic change in the subsynaptic reticulum (SSR) in the akt mutant occurred independent of stimulation. These results suggest that Akt functions in both pre- and postsynaptic growth and differentiation, and in particular, presynaptic action occurs in an activity-dependent manner.
Collapse
Affiliation(s)
- Yoon-Jung Kim
- Department of Physiology and Neuroscience, Dental Research Institute, Seoul National University School of Dentistry, Seoul, 03080, South Korea.
| |
Collapse
|
13
|
Xu KK, Yan Y, Yan SY, Xia PL, Yang WJ, Li C, Yang H. Disruption of the Serine/Threonine Kinase Akt Gene Affects Ovarian Development and Fecundity in the Cigarette Beetle, Lasioderma serricorne. Front Physiol 2021; 12:765819. [PMID: 34690822 PMCID: PMC8529032 DOI: 10.3389/fphys.2021.765819] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 09/16/2021] [Indexed: 11/17/2022] Open
Abstract
Serine/threonine kinase Akt, an important component of the insulin signaling pathway, plays an essential role in many physiological processes. In this study, we identified and characterized an Akt gene (designated LsAkt) from the cigarette beetle, Lasioderma serricorne. LsAkt contains a 1614 bp open reading frame encoding a 537 amino acid protein that possesses a conserved pleckstrin homology domain and a serine/threonine kinase domain. The expression of LsAkt was high in pupal stages and peaked in day-4 female pupae. In adult tissues, LsAkt was highly expressed in the thorax, ovary, and midgut. The expression of LsAkt was induced by methoprene or bovine insulin in vivo, but significantly decreased by 20-hydroxyecdysone. RNA interference-mediated knockdown of LsAkt resulted in severely blocked ovarian development and reduced fecundity and hatchability. The vitellogenin (Vg) content and juvenile hormone (JH) titers of LsAkt-depletion beetles were decreased, and expressions of Vg and four JH signaling and biosynthetic genes were significantly decreased. Silencing of LsAkt reduced the amounts of glucose, glycogen, and trehalose in female adults and affected the expressions of seven key carbohydrate metabolic genes. Taken together, it is inferred that Akt implicates in L. serricorne reproduction by modification of Vg synthesis, juvenile hormone production and carbohydrate metabolism.
Collapse
Affiliation(s)
- Kang-Kang Xu
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China.,Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, China
| | - Yi Yan
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China.,Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, China
| | - Shu-Yan Yan
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, China
| | | | - Wen-Jia Yang
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, China
| | - Can Li
- Guizhou Provincial Key Laboratory for Rare Animal and Economic Insect of the Mountainous Region, College of Biology and Environmental Engineering, Guiyang University, Guiyang, China
| | - Hong Yang
- Guizhou Provincial Key Laboratory for Agricultural Pest Management of Mountainous Region, Institute of Entomology, Guizhou University, Guiyang, China.,College of Tobacco Science, Guizhou University, Guiyang, China
| |
Collapse
|
14
|
Khan MM, Khan AH, Ali MW, Hafeez M, Ali S, Du C, Fan Z, Sattar M, Hua H. Emamectin benzoate induced enzymatic and transcriptional alternation in detoxification mechanism of predatory beetle Paederus fuscipes (Coleoptera: Staphylinidae) at the sublethal concentration. ECOTOXICOLOGY (LONDON, ENGLAND) 2021; 30:1227-1241. [PMID: 34117552 DOI: 10.1007/s10646-021-02426-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/17/2021] [Indexed: 06/12/2023]
Abstract
In this study, the detoxification enzyme activity and the transcriptional profile changes in the second instar through RNA-sequencing technology due to emamectin benzoate (EB) were assessed. The cytochrome P450 monooxygenases (P450) enzyme activity was not altered by EB due to the change in concentration and exposure time in all treatments. The glutathione S-transferase (GST) enzyme was not considerably varying in all treatments, while exposure time significantly changed the enzyme activity. Results showed that the esterase (Ests) activity was elevated with the increasing concentrations and exposure time. Two libraries were generated, containing 107,767,542 and 108,142,289 clean reads for the samples treated with LC30 of EB and control. These reads were grouped into 218,070 transcripts and 38,097 unigenes. A total of 2257 differentially expressed genes (DEGs) were identified from these unigenes, of which 599 up-regulated and 1658 were down-regulated. The majority of these DEGs related to pesticides resistance were identified in numerous Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, e.g., steroid hormone biosynthesis, glutathione metabolism, drug metabolism-other enzymes, chemical carcinogenesis, pathways of cancer, metabolism of xenobiotics by cytochrome P450, drug metabolism of cytochrome P450, linoleic acid metabolism, retinol metabolism, and insect hormone biosynthesis. These pathways also shared the same genes as cytochrome P450 monooxygenases (P450s), glutathione S-transferases (GSTs), Esterase (Ests), UDP-glucosyltransferases (UGTs), and ATP-binding cassettes (ABCs). A heatmap analysis also showed that regulation of genes in a pathway causes a series of gene expression regulation in subsequent pathways. Our quantitative reverse transcription-PCR (qRT-PCR) results were consistent with the DEG's data of transcriptome analysis. The comprehensive transcriptome sequence resource attained through this study evidence that the EB induces significant modification in enzyme activity and transcriptome profile of Paederus fuscipes, which may enable more significant molecular underpinnings behind the insecticide-resistance mechanism for further investigations.
Collapse
Affiliation(s)
- Muhammad Musa Khan
- Key Laboratory of Bio-Pesticide Innovation and Application, Engineering Research Centre of Biological Control, South China Agricultural University, Guangzhou, P.R. China.
| | - Aamir Hamid Khan
- National Key laboratory of crop genetic improvement, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| | - Muhammad Waqar Ali
- Institute of Fruit and Tea, Hubei Academy of Agricultural Sciences, Wuhan, P.R. China
| | - Muhammad Hafeez
- State Key Laboratory Breeding Base for Zhejiang Sustainable Pest and Disease Control, Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, P.R. China
| | - Shahbaz Ali
- Fareed Biodiversity and Conservation Centre, Department of Agricultural Engineering, Khawaja Fareed University of Engineering and Information Technology, Rahim Yar Khan, Punjab, Pakistan
| | - Cailian Du
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, P.R. China
| | - Zeyun Fan
- Key Laboratory of Bio-Pesticide Innovation and Application, Engineering Research Centre of Biological Control, South China Agricultural University, Guangzhou, P.R. China
| | - Muzammil Sattar
- Plant Protection Division, Nuclear Institute for Agriculture and Biology, Faisalabad, Punjab, Pakistan
| | - Hongxia Hua
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, College of Plant Science and Technology, Huazhong Agricultural University, Wuhan, P.R. China
| |
Collapse
|
15
|
Heier C, Klishch S, Stilbytska O, Semaniuk U, Lushchak O. The Drosophila model to interrogate triacylglycerol biology. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158924. [PMID: 33716135 DOI: 10.1016/j.bbalip.2021.158924] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 02/24/2021] [Accepted: 03/05/2021] [Indexed: 12/21/2022]
Abstract
The deposition of storage fat in the form of triacylglycerol (TAG) is an evolutionarily conserved strategy to cope with fluctuations in energy availability and metabolic stress. Organismal TAG storage in specialized adipose tissues provides animals a metabolic reserve that sustains survival during development and starvation. On the other hand, excessive accumulation of adipose TAG, defined as obesity, is associated with an increasing prevalence of human metabolic diseases. During the past decade, the fruit fly Drosophila melanogaster, traditionally used in genetics and developmental biology, has been established as a versatile model system to study TAG metabolism and the etiology of lipid-associated metabolic diseases. Similar to humans, Drosophila TAG homeostasis relies on the interplay of organ systems specialized in lipid uptake, synthesis, and processing, which are integrated by an endocrine network of hormones and messenger molecules. Enzymatic formation of TAG from sugar or dietary lipid, its storage in lipid droplets, and its mobilization by lipolysis occur via mechanisms largely conserved between Drosophila and humans. Notably, dysfunctional Drosophila TAG homeostasis occurs in the context of aging, overnutrition, or defective gene function, and entails tissue-specific and organismal pathologies that resemble human disease. In this review, we summarize the physiology and biochemistry of TAG in Drosophila and outline the potential of this organism as a model system to understand the genetic and dietary basis of TAG storage and TAG-related metabolic disorders.
Collapse
Affiliation(s)
- Christoph Heier
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Humboldtstrasse 50, A-8010 Graz, Austria; BioTechMed-Graz, Graz, Austria.
| | - Svitlana Klishch
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Olha Stilbytska
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Uliana Semaniuk
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine
| | - Oleh Lushchak
- Department of Biochemistry and Biotechnology, Department Biochemistry 1, Faculty of Natural Sciences, Vasyl Stefanyk Precarpathian National University, 57 Shevchenka str, Ivano-Frankivsk 76018, Ukraine.
| |
Collapse
|
16
|
Millington JW, Brownrigg GP, Basner-Collins PJ, Sun Z, Rideout EJ. Genetic manipulation of insulin/insulin-like growth factor signaling pathway activity has sex-biased effects on Drosophila body size. G3 (BETHESDA, MD.) 2021; 11:jkaa067. [PMID: 33793746 PMCID: PMC8063079 DOI: 10.1093/g3journal/jkaa067] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/09/2020] [Indexed: 12/14/2022]
Abstract
In Drosophila raised in nutrient-rich conditions, female body size is approximately 30% larger than male body size due to an increased rate of growth and differential weight loss during the larval period. While the mechanisms that control this sex difference in body size remain incompletely understood, recent studies suggest that the insulin/insulin-like growth factor signaling pathway (IIS) plays a role in the sex-specific regulation of processes that influence body size during development. In larvae, IIS activity differs between the sexes, and there is evidence of sex-specific regulation of IIS ligands. Yet, we lack knowledge of how changes to IIS activity impact body size in each sex, as the majority of studies on IIS and body size use single- or mixed-sex groups of larvae and/or adult flies. The goal of our current study was to clarify the body size requirement for IIS activity in each sex. To achieve this goal, we used established genetic approaches to enhance, or inhibit, IIS activity, and quantified pupal size in males and females. Overall, genotypes that inhibited IIS activity caused a female-biased decrease in body size, whereas genotypes that augmented IIS activity caused a male-specific increase in body size. These data extend our current understanding of body size regulation by showing that most changes to IIS pathway activity have sex-biased effects, and highlights the importance of analyzing body size data according to sex.
Collapse
Affiliation(s)
- Jason W Millington
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - George P Brownrigg
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Paige J Basner-Collins
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ziwei Sun
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Elizabeth J Rideout
- Department of Cellular and Physiological Sciences, Life Sciences Institute, The University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
17
|
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: 25] [Impact Index Per Article: 8.3] [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
|
18
|
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: 1.7] [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
|
19
|
Xu D, Yang H, Zhuo Z, Lu B, Hu J, Yang F. Characterization and analysis of the transcriptome in Opisina arenosella from different developmental stages using single-molecule real-time transcript sequencing and RNA-seq. Int J Biol Macromol 2020; 169:216-227. [PMID: 33340629 DOI: 10.1016/j.ijbiomac.2020.12.098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/10/2020] [Accepted: 12/12/2020] [Indexed: 02/06/2023]
Abstract
Opisina arenosella is one of the main pests harming coconut trees. To date, there have been few studies on the molecular genetics, biochemistry and physiology of O. arenosella at the transcriptional level, and there are no available reference genomes. Here, Illumina RNA sequencing combined with PacBio single-molecule real-time analysis was applied to study the transcriptome of this pest at different developmental stages, providing reference data for transcript expression analysis. Twelve samples of O. arenosella from different stages of development were sequenced using Illumina RNA sequencing, and the pooled RNA samples were sequenced with PacBio technology (Iso-Seq). A full-length transcriptome with 41,938 transcripts was captured, and the N50 and N90 lengths were 3543 bp and 1646 bp, respectively. A total of 36,925 transcripts were annotated in public databases, 6493 of which were long noncoding RNAs, while 2510 represented alternative splicing events. There were significant differences in the gene expression profiles at different developmental stages, with high levels of differential gene expression associated with growth, development, carbohydrate metabolism and immunity. This work provides resources and information for the study of the transcriptome and gene function of O. arenosella and provides a valuable foundation for understanding the changes in gene expression during development.
Collapse
Affiliation(s)
- Danping Xu
- College of Life Science, China West Normal University, Nanchong 637002, China
| | - Hongjun Yang
- College of Forestry, Hainan University, Haikou 570228, China
| | - Zhihang Zhuo
- College of Life Science, China West Normal University, Nanchong 637002, China; College of Forestry, Hainan University, Haikou 570228, China.
| | - Baoqian Lu
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Jiameng Hu
- College of Forestry, Hainan University, Haikou 570228, China
| | - Fan Yang
- College of Forestry, Hainan University, Haikou 570228, China
| |
Collapse
|
20
|
Meep, a Novel Regulator of Insulin Signaling, Supports Development and Insulin Sensitivity via Maintenance of Protein Homeostasis in Drosophila melanogaster. G3-GENES GENOMES GENETICS 2020; 10:4399-4410. [PMID: 32998936 PMCID: PMC7718763 DOI: 10.1534/g3.120.401688] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Insulin signaling is critical for developmental growth and adult homeostasis, yet the downstream regulators of this signaling pathway are not completely understood. Using the model organism Drosophila melanogaster, we took a genomic approach to identify novel mediators of insulin signaling. These studies led to the identification of Meep, encoded by the gene CG32335. Expression of this gene is both insulin receptor- and diet-dependent. We found that Meep was specifically required in the developing fat body to tolerate a high-sugar diet (HSD). Meep is not essential on a control diet, but when reared on an HSD, knockdown of meep causes hyperglycemia, reduced growth, developmental delay, pupal lethality, and reduced longevity. These phenotypes stem in part from Meep’s role in promoting insulin sensitivity and protein stability. This work suggests a critical role for protein homeostasis in development during overnutrition. Because Meep is conserved and obesity-associated in mammals, future studies on Meep may help to understand the role of proteostasis in insulin-resistant type 2 diabetes.
Collapse
|
21
|
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: 62] [Impact Index Per Article: 15.5] [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
|
22
|
Maity S, Das F, Kasinath BS, Ghosh-Choudhury N, Ghosh Choudhury G. TGFβ acts through PDGFRβ to activate mTORC1 via the Akt/PRAS40 axis and causes glomerular mesangial cell hypertrophy and matrix protein expression. J Biol Chem 2020; 295:14262-14278. [PMID: 32732288 DOI: 10.1074/jbc.ra120.014994] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/27/2020] [Indexed: 12/21/2022] Open
Abstract
Interaction of transforming growth factor-β (TGFβ)-induced canonical signaling with the noncanonical kinase cascades regulates glomerular hypertrophy and matrix protein deposition, which are early features of glomerulosclerosis. However, the specific target downstream of the TGFβ receptor involved in the noncanonical signaling is unknown. Here, we show that TGFβ increased the catalytic loop phosphorylation of platelet-derived growth factor receptor β (PDGFRβ), a receptor tyrosine kinase expressed abundantly in glomerular mesangial cells. TGFβ increased phosphorylation of the PI 3-kinase-interacting Tyr-751 residue of PDGFRβ, thus activating Akt. Inhibition of PDGFRβ using a pharmacological inhibitor and siRNAs blocked TGFβ-stimulated phosphorylation of proline-rich Akt substrate of 40 kDa (PRAS40), an intrinsic inhibitory component of mTORC1, and prevented activation of mTORC1 in the absence of any effect on Smad 2/3 phosphorylation. Expression of constitutively active myristoylated Akt reversed the siPDGFRβ-mediated inhibition of mTORC1 activity; however, co-expression of the phospho-deficient mutant of PRAS40 inhibited the effect of myristoylated Akt, suggesting a definitive role of PRAS40 phosphorylation in mTORC1 activation downstream of PDGFRβ in mesangial cells. Additionally, we demonstrate that PDGFRβ-initiated phosphorylation of PRAS40 is required for TGFβ-induced mesangial cell hypertrophy and fibronectin and collagen I (α2) production. Increased activating phosphorylation of PDGFRβ is also associated with enhanced TGFβ expression and mTORC1 activation in the kidney cortex and glomeruli of diabetic mice and rats, respectively. Thus, pursuing TGFβ noncanonical signaling, we identified how TGFβ receptor I achieves mTORC1 activation through PDGFRβ-mediated Akt/PRAS40 phosphorylation to spur mesangial cell hypertrophy and matrix protein accumulation. These findings provide support for targeting PDGFRβ in TGFβ-driven renal fibrosis.
Collapse
Affiliation(s)
- Soumya Maity
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Falguni Das
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Balakuntalam S Kasinath
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA.,Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| | | | - Goutam Ghosh Choudhury
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas, USA .,Department of Veterans Affairs Research, South Texas Veterans Health Care System, San Antonio, Texas, USA.,Geriatric Research, Education, and Clinical Center, South Texas Veterans Health Care System, San Antonio, Texas, USA
| |
Collapse
|
23
|
Ho CH, Treisman JE. Specific Isoforms of the Guanine-Nucleotide Exchange Factor dPix Couple Neuromuscular Synapse Growth to Muscle Growth. Dev Cell 2020; 54:117-131.e5. [PMID: 32516570 DOI: 10.1016/j.devcel.2020.05.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/09/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022]
Abstract
Developmental growth requires coordination between the growth rates of individual tissues and organs. Here, we examine how Drosophila neuromuscular synapses grow to match the size of their target muscles. We show that changes in muscle growth driven by autonomous modulation of insulin receptor signaling produce corresponding changes in synapse size, with each muscle affecting only its presynaptic motor neuron branches. This scaling growth is mechanistically distinct from synaptic plasticity driven by neuronal activity and requires increased postsynaptic differentiation induced by insulin receptor signaling in muscle. We identify the guanine-nucleotide exchange factor dPix as an effector of insulin receptor signaling. Alternatively spliced dPix isoforms that contain a specific exon are necessary and sufficient for postsynaptic differentiation and scaling growth, and their mRNA levels are regulated by insulin receptor signaling. These findings define a mechanism by which the same signaling pathway promotes both autonomous muscle growth and non-autonomous synapse growth.
Collapse
Affiliation(s)
- Cheuk Hei Ho
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
24
|
Poe AR, Xu Y, Zhang C, Lei J, Li K, Labib D, Han C. Low FoxO expression in Drosophila somatosensory neurons protects dendrite growth under nutrient restriction. eLife 2020; 9:53351. [PMID: 32427101 PMCID: PMC7308081 DOI: 10.7554/elife.53351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/18/2020] [Indexed: 12/22/2022] Open
Abstract
During prolonged nutrient restriction, developing animals redistribute vital nutrients to favor brain growth at the expense of other organs. In Drosophila, such brain sparing relies on a glia-derived growth factor to sustain proliferation of neural stem cells. However, whether other aspects of neural development are also spared under nutrient restriction is unknown. Here we show that dynamically growing somatosensory neurons in the Drosophila peripheral nervous system exhibit organ sparing at the level of arbor growth: Under nutrient stress, sensory dendrites preferentially grow as compared to neighboring non-neural tissues, resulting in dendrite overgrowth. These neurons express lower levels of the stress sensor FoxO than neighboring epidermal cells, and hence exhibit no marked induction of autophagy and a milder suppression of Tor signaling under nutrient stress. Preferential dendrite growth allows for heightened animal responses to sensory stimuli, indicative of a potential survival advantage under environmental challenges.
Collapse
Affiliation(s)
- Amy R Poe
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Yineng Xu
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Christine Zhang
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Joyce Lei
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Kailyn Li
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - David Labib
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| | - Chun Han
- Weill Institute for Cell and Molecular Biology and Department of Molecular Biology and Genetics, Cornell University, Ithaca, United States
| |
Collapse
|
25
|
Toprak U. The Role of Peptide Hormones in Insect Lipid Metabolism. Front Physiol 2020; 11:434. [PMID: 32457651 PMCID: PMC7221030 DOI: 10.3389/fphys.2020.00434] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Lipids are the primary storage molecules and an essential source of energy in insects during reproduction, prolonged periods of flight, starvation, and diapause. The coordination center for insect lipid metabolism is the fat body, which is analogous to the vertebrate adipose tissue and liver. The fat body is primarily composed of adipocytes, which accumulate triacylglycerols in intracellular lipid droplets. Genomics and proteomics, together with functional analyses, such as RNA interference and CRISPR/Cas9-targeted genome editing, identified various genes involved in lipid metabolism and elucidated their functions. However, the endocrine control of insect lipid metabolism, in particular the roles of peptide hormones in lipogenesis and lipolysis are relatively less-known topics. In the current review, the neuropeptides that directly or indirectly affect insect lipid metabolism are introduced. The primary lipolytic and lipogenic peptide hormones are adipokinetic hormone and the brain insulin-like peptides (ILP2, ILP3, ILP5). Other neuropeptides, such as insulin-growth factor ILP6, neuropeptide F, allatostatin-A, corazonin, leucokinin, tachykinins and limostatin, might stimulate lipolysis, while diapause hormone-pheromone biosynthesis activating neuropeptide, short neuropeptide F, CCHamide-2, and the cytokines Unpaired 1 and Unpaired 2 might induce lipogenesis. Most of these peptides interact with one another, but mostly with insulin signaling, and therefore affect lipid metabolism indirectly. Peptide hormones are also involved in lipid metabolism during reproduction, flight, diapause, starvation, infections and immunity; these are also highlighted. The review concludes with a discussion of the potential of lipid metabolism-related peptide hormones in pest management.
Collapse
Affiliation(s)
- Umut Toprak
- Molecular Entomology Lab., Department of Plant Protection Ankara, Faculty of Agriculture, Ankara University, Ankara, Turkey
| |
Collapse
|
26
|
Sudhakar SR, Pathak H, Rehman N, Fernandes J, Vishnu S, Varghese J. Insulin signalling elicits hunger-induced feeding in Drosophila. Dev Biol 2020; 459:87-99. [DOI: 10.1016/j.ydbio.2019.11.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/21/2022]
|
27
|
Kim M, Sujkowski A, Namkoong S, Gu B, Cobb T, Kim B, Kowalsky AH, Cho CS, Semple I, Ro SH, Davis C, Brooks SV, Karin M, Wessells RJ, Lee JH. Sestrins are evolutionarily conserved mediators of exercise benefits. Nat Commun 2020; 11:190. [PMID: 31929512 PMCID: PMC6955242 DOI: 10.1038/s41467-019-13442-5] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 11/06/2019] [Indexed: 01/04/2023] Open
Abstract
Exercise is among the most effective interventions for age-associated mobility decline and metabolic dysregulation. Although long-term endurance exercise promotes insulin sensitivity and expands respiratory capacity, genetic components and pathways mediating the metabolic benefits of exercise have remained elusive. Here, we show that Sestrins, a family of evolutionarily conserved exercise-inducible proteins, are critical mediators of exercise benefits. In both fly and mouse models, genetic ablation of Sestrins prevents organisms from acquiring metabolic benefits of exercise and improving their endurance through training. Conversely, Sestrin upregulation mimics both molecular and physiological effects of exercise, suggesting that it could be a major effector of exercise metabolism. Among the various targets modulated by Sestrin in response to exercise, AKT and PGC1α are critical for the Sestrin effects in extending endurance. These results indicate that Sestrin is a key integrating factor that drives the benefits of chronic exercise to metabolism and physical endurance.
Collapse
Affiliation(s)
- Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Alyson Sujkowski
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Sim Namkoong
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon, 24341, Republic of Korea
| | - Bondong Gu
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tyler Cobb
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Boyoung Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Allison H Kowalsky
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Chun-Seok Cho
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Ian Semple
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Seung-Hyun Ro
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
- Department of Biochemistry, University of Nebraska, Lincoln, NE, 68588, USA
| | - Carol Davis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Susan V Brooks
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Michael Karin
- Department of Pharmacology, University of California San Diego, La Jolla, CA, 92093, USA
| | - Robert J Wessells
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, 48201, USA.
| | - Jun Hee Lee
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
28
|
Tu T, Chen J, Chen L, Stiles BL. Dual-Specific Protein and Lipid Phosphatase PTEN and Its Biological Functions. Cold Spring Harb Perspect Med 2020; 10:cshperspect.a036301. [PMID: 31548229 DOI: 10.1101/cshperspect.a036301] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Phosphatase and tensin homolog deleted on chromosome 10 (PTEN) encodes a 403-amino acid protein with an amino-terminal domain that shares sequence homology with the actin-binding protein tensin and the putative tyrosine-protein phosphatase auxilin. Crystal structure analysis of PTEN has revealed a C2 domain that binds to phospholipids in membranes and a phosphatase domain that displays dual-specific activity toward both tyrosine (Y), serine (S)/threonine (T), as well as lipid substrates in vitro. Characterized primarily as a lipid phosphatase, PTEN plays important roles in multiple cellular processes including cell growth/survival as well as metabolism.
Collapse
Affiliation(s)
- Taojian Tu
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Jingyu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Lulu Chen
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA
| | - Bangyan L Stiles
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California 90033, USA.,Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, California 90033, USA
| |
Collapse
|
29
|
Lin X, Smagghe G. Roles of the insulin signaling pathway in insect development and organ growth. Peptides 2019; 122:169923. [PMID: 29458057 DOI: 10.1016/j.peptides.2018.02.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 12/16/2022]
Abstract
Organismal development is a complex process as it requires coordination of many aspects to grow into fit individuals, such as the control of body size and organ growth. Therefore, the mechanisms of precise control of growth are essential for ensuring the growth of organisms at a correct body size and proper organ proportions during development. The control of the growth rate and the duration of growth (or the cessation of growth) are required in size control. The insulin signaling pathway and the elements involved are essential in the control of growth. On the other hand, the ecdysteroid molting hormone determines the duration of growth. The secretion of these hormones is controlled by environmental factors such as nutrition. Moreover, the target of rapamycin (TOR) pathway is considered as a nutrient sensing pathway. Important cross-talks have been shown to exist among these pathways. In this review, we outline the control of body and organ growth by the insulin/TOR signaling pathway, and also the interaction between nutrition via insulin/TOR signaling and ecdysteroids at the coordination of organismal development and organ growth in insects, mainly focusing on the well-studied fruit fly Drosophila melanogaster.
Collapse
Affiliation(s)
- Xianyu Lin
- Department of Crop Protection, Ghent University, 9000 Ghent, Belgium
| | - Guy Smagghe
- Department of Crop Protection, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
30
|
Borreguero-Muñoz N, Fletcher GC, Aguilar-Aragon M, Elbediwy A, Vincent-Mistiaen ZI, Thompson BJ. The Hippo pathway integrates PI3K-Akt signals with mechanical and polarity cues to control tissue growth. PLoS Biol 2019; 17:e3000509. [PMID: 31613895 PMCID: PMC6814241 DOI: 10.1371/journal.pbio.3000509] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 10/25/2019] [Accepted: 10/03/2019] [Indexed: 11/19/2022] Open
Abstract
The Hippo signalling pathway restricts cell proliferation in animal tissues by inhibiting Yes-associated protein (YAP or YAP1) and Transcriptional Activator with a PDZ domain (TAZ or WW-domain-containing transcriptional activator [WWTR1]), coactivators of the Scalloped (Sd or TEAD) DNA-binding transcription factor. Drosophila has a single YAP/TAZ homolog named Yorkie (Yki) that is regulated by Hippo pathway signalling in response to epithelial polarity and tissue mechanics during development. Here, we show that Yki translocates to the nucleus to drive Sd-mediated cell proliferation in the ovarian follicle cell epithelium in response to mechanical stretching caused by the growth of the germline. Importantly, mechanically induced Yki nuclear localisation also requires nutritionally induced insulin/insulin-like growth factor 1 (IGF-1) signalling (IIS) via phosphatidyl inositol-3-kinase (PI3K), phosphoinositide-dependent kinase 1 (PDK1 or PDPK1), and protein kinase B (Akt or PKB) in the follicular epithelium. We find similar results in the developing Drosophila wing, where Yki becomes nuclear in the mechanically stretched cells of the wing pouch during larval feeding, which induces IIS, but translocates to the cytoplasm upon cessation of feeding in the third instar stage. Inactivating Akt prevents nuclear Yki localisation in the wing disc, while ectopic activation of the insulin receptor, PI3K, or Akt/PKB is sufficient to maintain nuclear Yki in mechanically stimulated cells of the wing pouch even after feeding ceases. Finally, IIS also promotes YAP nuclear localisation in response to mechanical cues in mammalian skin epithelia. Thus, the Hippo pathway has a physiological function as an integrator of epithelial cell polarity, tissue mechanics, and nutritional cues to control cell proliferation and tissue growth in both Drosophila and mammals.
Collapse
Affiliation(s)
| | - Georgina C. Fletcher
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Mario Aguilar-Aragon
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Ahmed Elbediwy
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
| | | | - Barry J. Thompson
- Epithelial Biology Laboratory, The Francis Crick Institute, London, United Kingdom
- EMBL Australia, Department of Cancer Biology & Therapeutics, The John Curtin School of Medical Research, The Australian National University, Acton, Australia
- * E-mail:
| |
Collapse
|
31
|
Suzawa M, Muhammad NM, Joseph BS, Bland ML. The Toll Signaling Pathway Targets the Insulin-like Peptide Dilp6 to Inhibit Growth in Drosophila. Cell Rep 2019; 28:1439-1446.e5. [DOI: 10.1016/j.celrep.2019.07.015] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/28/2019] [Accepted: 07/03/2019] [Indexed: 01/08/2023] Open
|
32
|
Lin X, De Schutter K, Chafino S, Franch-Marro X, Martín D, Smagghe G. Target of rapamycin (TOR) determines appendage size during pupa formation of the red flour beetle Tribolium castaneum. JOURNAL OF INSECT PHYSIOLOGY 2019; 117:103902. [PMID: 31233769 DOI: 10.1016/j.jinsphys.2019.103902] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 06/19/2019] [Accepted: 06/20/2019] [Indexed: 06/09/2023]
Abstract
The adult body size is species-specific and controlled by complex interactions between hormones and the IIS/TOR pathway. To analyze the role of target of rapamycin (TOR) in the growth and development of the insect, expression levels of TOR were silenced in the model and pest insect red flour beetle, Tribolium castaneum. Injection of dsRNA into the last larval instar decreased pupal mass and size, while the amount of food intake by the larvae was not affected. These results place TcTOR downstream of nutrition as a transducer for nutritional signals to increase larval growth. In addition, TcTOR-silencing notably decreased the size of the adult appendages. Analysis of the wings and elytra revealed a decrease in cell size and number of these appendages in the TcTOR-silenced insects. This reduction in size was correlated with a decrease of transcriptional levels of marker genes controlling the cell cycle. Altogether, these results suggest a pivotal role for TcTOR in integrating nutritional signals and regulation of body and appendages growth.
Collapse
Affiliation(s)
- Xianyu Lin
- Department of Plants and Crops, Ghent University, 9000 Ghent, Belgium
| | | | - Silvia Chafino
- Institute of Evolutionary Biology (IBE), CSIC- Pompeu Fabra University, E-08003 Barcelona, Spain
| | - Xavier Franch-Marro
- Institute of Evolutionary Biology (IBE), CSIC- Pompeu Fabra University, E-08003 Barcelona, Spain
| | - David Martín
- Institute of Evolutionary Biology (IBE), CSIC- Pompeu Fabra University, E-08003 Barcelona, Spain
| | - Guy Smagghe
- Department of Plants and Crops, Ghent University, 9000 Ghent, Belgium.
| |
Collapse
|
33
|
Pletcher RC, Hardman SL, Intagliata SF, Lawson RL, Page A, Tennessen JM. A Genetic Screen Using the Drosophila melanogaster TRiP RNAi Collection To Identify Metabolic Enzymes Required for Eye Development. G3 (BETHESDA, MD.) 2019; 9:2061-2070. [PMID: 31036678 PMCID: PMC6643872 DOI: 10.1534/g3.119.400193] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 04/26/2019] [Indexed: 01/05/2023]
Abstract
The metabolic enzymes that compose glycolysis, the citric acid cycle, and other pathways within central carbon metabolism have emerged as key regulators of animal development. These enzymes not only generate the energy and biosynthetic precursors required to support cell proliferation and differentiation, but also moonlight as regulators of transcription, translation, and signal transduction. Many of the genes associated with animal metabolism, however, have never been analyzed in a developmental context, thus highlighting how little is known about the intersection of metabolism and development. Here we address this deficiency by using the Drosophila TRiP RNAi collection to disrupt the expression of over 1,100 metabolism-associated genes within cells of the eye imaginal disc. Our screen not only confirmed previous observations that oxidative phosphorylation serves a critical role in the developing eye, but also implicated a host of other metabolic enzymes in the growth and differentiation of this organ. Notably, our analysis revealed a requirement for glutamine and glutamate metabolic processes in eye development, thereby revealing a role of these amino acids in promoting Drosophila tissue growth. Overall, our analysis highlights how the Drosophila eye can serve as a powerful tool for dissecting the relationship between development and metabolism.
Collapse
Affiliation(s)
- Rose C Pletcher
- Department of Biology, Indiana University, 1001 East Third Street, Bloomington, IN 47405
| | - Sara L Hardman
- Department of Biology, Indiana University, 1001 East Third Street, Bloomington, IN 47405
| | - Sydney F Intagliata
- Department of Biology, Indiana University, 1001 East Third Street, Bloomington, IN 47405
| | - Rachael L Lawson
- Department of Biology, Indiana University, 1001 East Third Street, Bloomington, IN 47405
| | - Aumunique Page
- Department of Biology, Indiana University, 1001 East Third Street, Bloomington, IN 47405
| | - Jason M Tennessen
- Department of Biology, Indiana University, 1001 East Third Street, Bloomington, IN 47405
| |
Collapse
|
34
|
Bowling S, Lawlor K, Rodríguez TA. Cell competition: the winners and losers of fitness selection. Development 2019; 146:146/13/dev167486. [PMID: 31278123 DOI: 10.1242/dev.167486] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The process of cell competition results in the 'elimination of cells that are viable but less fit than surrounding cells'. Given the highly heterogeneous nature of our tissues, it seems increasingly likely that cells are engaged in a 'survival of the fittest' battle throughout life. The process has a myriad of positive roles in the organism: it selects against mutant cells in developing tissues, prevents the propagation of oncogenic cells and eliminates damaged cells during ageing. However, 'super-fit' cancer cells can exploit cell competition mechanisms to expand and spread. Here, we review the regulation, roles and risks of cell competition in organism development, ageing and disease.
Collapse
Affiliation(s)
- Sarah Bowling
- National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Katerina Lawlor
- National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Tristan A Rodríguez
- National Heart and Lung Institute, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
35
|
Urwyler O, Izadifar A, Vandenbogaerde S, Sachse S, Misbaer A, Schmucker D. Branch-restricted localization of phosphatase Prl-1 specifies axonal synaptogenesis domains. Science 2019; 364:364/6439/eaau9952. [DOI: 10.1126/science.aau9952] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 03/29/2019] [Indexed: 01/21/2023]
Abstract
Central nervous system (CNS) circuit development requires subcellular control of synapse formation and patterning of synapse abundance. We identified the Drosophila membrane-anchored phosphatase of regenerating liver (Prl-1) as an axon-intrinsic factor that promotes synapse formation in a spatially restricted fashion. The loss of Prl-1 in mechanosensory neurons reduced the number of CNS presynapses localized on a single axon collateral and organized as a terminal arbor. Flies lacking all Prl-1 protein had locomotor defects. The overexpression of Prl-1 induced ectopic synapses. In mechanosensory neurons, Prl-1 modulates the insulin receptor (InR) signaling pathway within a single contralateral axon compartment, thereby affecting the number of synapses. The axon branch–specific localization and function of Prl-1 depend on untranslated regions of the prl-1 messenger RNA (mRNA). Therefore, compartmentalized restriction of Prl-1 serves as a specificity factor for the subcellular control of axonal synaptogenesis.
Collapse
|
36
|
Chang BH, Cui B, Ullah H, Li S, Hao K, Tu X, Wang G, Nong X, McNeill MR, Huang X, Zhang Z. Role of PTP/PTK trans activated insulin-like signalling pathway in regulation of grasshopper (Oedaleus asiaticus) development. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:8312-8324. [PMID: 30706274 DOI: 10.1007/s11356-019-04212-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/08/2019] [Indexed: 06/09/2023]
Abstract
Protein tyrosine phosphatase (PTPs) and protein tyrosine kinase (PTKs) genes are responsible for the regulation of insect insulin-like pathway (ILP), cells growth, metabolism initiation, gene transcription and observing immune response. Signal transduction in insect cell is also associated with PTPs and PTKs. The grasshopper (Oedaleus asiaticus) 'Bey-Bienko' were treated with dsRNA of protein tyrosine non-receptor type 4 (PTPN4) and protein tyrosine kinase 5 (PTK5) along with control (water). Applying dsPTK5 treatments in 5th instar of Oedaleus asiaticus, significant reduction was recorded in body dry mass, growth rate and overall performance except survival rate. Whereas with PTPN4, no such significant impact on all of these growth parameters was recorded. Expression of genes in ILP 5th instar of Oedaleus asiaticus by the application of dsPTPN4 and dsPTK5 revealed that PTK, INSR (insulin receptor), IRS (insulin receptor substrate), PI3K (phosphoinositide 3-kinase), PDK (3-phosphoinositide-dependent protein kinase), Akt (protein kinase B) and FOXO (forkhead transcription factor) significantly expressed with downregulation except PTPN4, which remained non-significant. On the other hand, the phosphorylation level of ILP four proteins in O. asiaticus with the treatment of dsPTPN4 and dsPTK5 significantly affected P-IRS and P-FOXO, while P-INSR and P-AKT remained stable at the probability level of 5%. This indicated that the stress response in the O. asiaticus insulin-like signalling pathway (ILP) reduced. Regarding association of protective enzymatic activities, ROS (relative oxygen species), CAT (catalase) and PO (phenol oxidase) increased significantly with exposure to dsPTK5 as compared to dsPTPN4 and control, while exposure of 5th instar of O. asiaticus to dsPTPN4 treatment slightly raised CAT and PO activities with but significant contribution. No such significant effect on MFO and POD was seen using dsPTPN4 and dsPTK5. This showed that in the ILP of O. asiaticus, PTK5 was detrimental to growth, body mass and overall performance, which ultimately benefited insect detoxification with high-energy cost.
Collapse
Affiliation(s)
- Babar Hussain Chang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | - Boyang Cui
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | - Hidayat Ullah
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | - Shuang Li
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | - Kun Hao
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | - Xiongbing Tu
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | - Guangjun Wang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | - Xiangqun Nong
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China
| | | | - Xunbing Huang
- College of Agriculture and Forestry Science, Linyi University, Linyi, China.
| | - Zehua Zhang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection, Chinese Academy of Agricultural Science, Beijing, China.
- Scientific Observation and Experimental Station of Pests in Xilin Gol Rangeland, Ministry of Agriculture and Rural Affairs, Xilinhot, China.
| |
Collapse
|
37
|
Triacylglycerol Metabolism in Drosophila melanogaster. Genetics 2019; 210:1163-1184. [PMID: 30523167 DOI: 10.1534/genetics.118.301583] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022] Open
Abstract
Triacylglycerol (TAG) is the most important caloric source with respect to energy homeostasis in animals. In addition to its evolutionarily conserved importance as an energy source, TAG turnover is crucial to the metabolism of structural and signaling lipids. These neutral lipids are also key players in development and disease. Here, we review the metabolism of TAG in the Drosophila model system. Recently, the fruit fly has attracted renewed attention in research due to the unique experimental approaches it affords in studying the tissue-autonomous and interorgan regulation of lipid metabolism in vivo Following an overview of the systemic control of fly body fat stores, we will cover lipid anabolic, enzymatic, and regulatory processes, which begin with the dietary lipid breakdown and de novo lipogenesis that results in lipid droplet storage. Next, we focus on lipolytic processes, which mobilize storage TAG to make it metabolically accessible as either an energy source or as a building block for biosynthesis of other lipid classes. Since the buildup and breakdown of fat involves various organs, we highlight avenues of lipid transport, which are at the heart of functional integration of organismic lipid metabolism. Finally, we draw attention to some "missing links" in basic neutral lipid metabolism and conclude with a perspective on how fly research can be exploited to study functional metabolic roles of diverse lipids.
Collapse
|
38
|
Deng P, Xu QY, Fu KY, Guo WC, Li GQ. RNA interference against the putative insulin receptor substrate gene chico affects metamorphosis in Leptinotarsa decemlineata. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2018; 103:1-11. [PMID: 30296480 DOI: 10.1016/j.ibmb.2018.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 09/30/2018] [Accepted: 10/05/2018] [Indexed: 06/08/2023]
Abstract
It is noted that insect insulin/insulin-like growth factor/target of rapamycin signaling is critical for the regulation of metamorphosis in holometabolous insects. However, the molecular mechanism remains undetermined. Our previous findings reveal that RNA interference (RNAi)-mediated knockdown of an insulin gene (LdILP2) in Leptinotarsa decemlineata disturbs both 20-hydroxyecdysone (20E) and juvenile hormone (JH) signaling, and impairs pupation. In the present paper, we further observed that the expression of the insulin receptor substrate gene chico (Ldchico) and the phosphoinositide-3-kinase gene pi3k (Ldpi3k92E) was repressed in LdILP2 depleted larvae. Moreover, RNAi of Ldchico or Ldpi3k92E decreased food consumption, affected absorption and metabolism of amino acids and sugars, and reduced expression of several 20E (LdEcR, LdHR3 and LdE75) and JH (LdJHAMT, LdKr-h1 and LdHairy) signaling genes. As a result, larval development was postponed and larval growth was inhibited. Intriguingly, knockdown of Ldchico, rather than Ldpi3k92E, impaired larval-pupal and pupal-adult ecdysis, and specifically repressed transcription of another 20E signaling gene LdUSP. Ingestion of 20E rescued the expression of LdEcR, LdHR3 and LdE75, whereas 20E feeding restored neither the decreased LdUSP mRNA level, nor the reduced pupation and adult emergence rates in Ldchico RNAi larvae. Therefore, Chico is critical for the regulation of larval-pupal-adult transition by a PI3K-independent pathway, perhaps through activation of USP in L. decemlineata.
Collapse
Affiliation(s)
- Pan Deng
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Qing-Yu Xu
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing, 210095, China.
| | - Kai-Yun Fu
- Department of Plant Protection, Xinjiang Academy of Agricultural Sciences, Urumqi, 830091, China.
| | - Wen-Chao Guo
- Department of Plant Protection, Xinjiang Academy of Agricultural Sciences, Urumqi, 830091, China; Xinjiang Laboratory of Special Environmental Microbiology, Institute of Microbiology, Xinjiang Academy of Agricultural Sciences, Urumqi, 830091, China.
| | - Guo-Qing Li
- Education Ministry Key Laboratory of Integrated Management of Crop Diseases and Pests, College of Plant Protection, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
39
|
Madan E, Gogna R, Moreno E. Cell competition in development: information from flies and vertebrates. Curr Opin Cell Biol 2018; 55:150-157. [DOI: 10.1016/j.ceb.2018.08.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/03/2018] [Accepted: 08/03/2018] [Indexed: 12/20/2022]
|
40
|
Akt2 causes TGFβ-induced deptor downregulation facilitating mTOR to drive podocyte hypertrophy and matrix protein expression. PLoS One 2018; 13:e0207285. [PMID: 30444896 PMCID: PMC6239304 DOI: 10.1371/journal.pone.0207285] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 10/29/2018] [Indexed: 02/06/2023] Open
Abstract
TGFβ promotes podocyte hypertrophy and expression of matrix proteins in fibrotic kidney diseases such as diabetic nephropathy. Both mTORC1 and mTORC2 are hyperactive in response to TGFβ in various renal diseases. Deptor is a component of mTOR complexes and a constitutive inhibitor of their activities. We identified that deptor downregulation by TGFβ maintains hyperactive mTOR in podocytes. To unravel the mechanism, we found that TGFβ -initiated noncanonical signaling controls deptor inhibition. Pharmacological inhibitor of PI 3 kinase, Ly 294002 and pan Akt kinase inhibitor MK 2206 prevented the TGFβ induced downregulation of deptor, resulting in suppression of both mTORC1 and mTORC2 activities. However, specific isoform of Akt involved in this process is not known. We identified Akt2 as predominant isoform expressed in kidney cortex, glomeruli and podocytes. TGFβ time-dependently increased the activating phosphorylation of Akt2. Expression of dominant negative PI 3 kinase and its signaling inhibitor PTEN blocked Akt2 phosphorylation by TGFβ. Inhibition of Akt2 using a phospho-deficient mutant that inactivates its kinase activity, as well as siRNA against the kinase markedly diminished TGFβ -mediated deptor suppression, its association with mTOR and activation of mTORC1 and mTORC2. Importantly, inhibition of Akt2 blocked TGFβ -induced podocyte hypertrophy and expression of the matrix protein fibronectin. This inhibition was reversed by the downregulation of deptor. Interestingly, we detected increased phosphorylation of Akt2 concomitant with TGFβ expression in the kidneys of diabetic rats. Thus, our data identify previously unrecognized Akt2 kinase as a driver of TGFβ induced deptor downregulation and sustained mTORC1 and mTORC2 activation. Furthermore, we provide the first evidence that deptor downstream of Akt2 contributes to podocyte hypertrophy and matrix protein expression found in glomerulosclerosis in different renal diseases.
Collapse
|
41
|
Serotonin signaling regulates insulin-like peptides for growth, reproduction, and metabolism in the disease vector Aedes aegypti. Proc Natl Acad Sci U S A 2018; 115:E9822-E9831. [PMID: 30275337 DOI: 10.1073/pnas.1808243115] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Disease-transmitting female mosquitoes require a vertebrate blood meal to produce their eggs. An obligatory hematophagous lifestyle, rapid reproduction, and existence of a large number of transmittable diseases make mosquitoes the world's deadliest animals. Attaining optimal body size and nutritional status is critical for mosquitoes to become reproductively competent and effective disease vectors. We report that blood feeding boosts serotonin concentration and elevates the serotonin receptor Aa5HT2B (Aedes aegypti 5-hydroxytryptamine receptor, type 2B) transcript level in the fat-body, an insect analog of the vertebrate liver and adipose tissue. Aa5HT2B gene disruption using the CRISPR-Cas9 gene-editing approach led to a decreased body size, postponed development, shortened lifespan, retarded ovarian growth, and dramatically diminished lipid accumulation. Expression of the insulin-like peptide (ILP) genes ilp2 and ilp6 was down-regulated while that of ilp5 and ilp4 was up-regulated in response to Aa5HT2B disruption. CRISPR-Cas9 disruption of ilp2 or ilp6 resulted in adverse phenotypes similar to those of Aa5HT2B disruption, while ilp5 CRISPR-Cas9 disruption had exactly the opposite effect on growth and metabolism, with significantly increased body size and elevated lipid stores. Simultaneous CRISPR-Cas9 disruption of Aa5HT2B and ilp5 rescued these phenotypic manifestations. Aa5HT2B RNAi silencing rendered ilp6 insensitive to serotonin treatment in the cultured fat-body, suggesting a regulatory link between Aa5HT2B and ILP6. Moreover, CRISPR-Cas9 ilp6 disruption affects expression of ilp-2, -5, and -4, pointing out on a possible role of ILP6 as a mediator of the Aa5HT2B action.
Collapse
|
42
|
Ahmad M, Keebaugh ES, Tariq M, Ja WW. Evolutionary responses of Drosophila melanogaster under chronic malnutrition. Front Ecol Evol 2018; 6. [PMID: 31286000 DOI: 10.3389/fevo.2018.00047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Drosophila species have successfully spread and adapted to diverse climates across the globe. For D. melanogaster, rotting vegetative matter provides the primary substrate for mating and oviposition, and also acts as a nutritional resource for developing larvae and adult flies. The transitory nature of decaying vegetation exposes D. melanogaster to rapidly changing nutrient availability. As evidenced by their successful global spread, flies are capable of dealing with fluctuating nutritional reserves within their respective ecological niches. Therefore, D. melanogaster populations might contain standing genetic variation to support survival during periods of nutrient scarcity. The natural history and genetic tractability of D. melanogaster make the fly an ideal model for studies on the genetic basis of resistance to nutritional stress. We review artificial selection studies on nutritionally-deprived D. melanogaster and summarize the phenotypic outcomes of selected animals. Many of the reported evolved traits phenocopy mutants of the nutrient-sensing PI3K/Akt pathway. Given that the PI3K/Akt pathway is also responsive to acute nutritional stress, the PI3K/Akt pathway might underlie traits evolved under chronic nutritional deprivation. Future studies that directly test for the genetic mechanisms driving evolutionary responses to nutritional stress will take advantage of the ease in manipulating fly nutrient availability in the laboratory.
Collapse
Affiliation(s)
- Muhammad Ahmad
- Department of Biology, SBA School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan.,Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA.,Center on Aging, The Scripps Research Institute, Jupiter, Florida, USA
| | - Erin S Keebaugh
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA.,Center on Aging, The Scripps Research Institute, Jupiter, Florida, USA
| | - Muhammad Tariq
- Department of Biology, SBA School of Science and Engineering, Lahore University of Management Sciences, Lahore, Pakistan
| | - William W Ja
- Department of Neuroscience, The Scripps Research Institute, Jupiter, Florida, USA.,Center on Aging, The Scripps Research Institute, Jupiter, Florida, USA
| |
Collapse
|
43
|
Thounaojam B, Keshan B. Modulation of gene expression by nutritional state and hormones in Bombyx larvae in relation to its growth period. Gene Expr Patterns 2017; 25-26:175-183. [DOI: 10.1016/j.gep.2017.08.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 08/05/2017] [Accepted: 08/22/2017] [Indexed: 10/19/2022]
|
44
|
Das F, Ghosh-Choudhury N, Kasinath BS, Choudhury GG. Tyrosines-740/751 of PDGFRβ contribute to the activation of Akt/Hif1α/TGFβ nexus to drive high glucose-induced glomerular mesangial cell hypertrophy. Cell Signal 2017; 42:44-53. [PMID: 28951244 DOI: 10.1016/j.cellsig.2017.09.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 09/22/2017] [Indexed: 01/24/2023]
Abstract
Glomerular mesangial cell hypertrophy contributes to the complications of diabetic nephropathy. The mechanism by which high glucose induces mesangial cell hypertrophy is poorly understood. Here we explored the role of the platelet-derived growth factor receptor-β (PDGFRβ) tyrosine kinase in driving the high glucose-induced mesangial cell hypertrophy. We show that high glucose stimulates the association of the PDGFRβ with PI 3 kinase leading to tyrosine phosphorylation of the latter. High glucose-induced Akt kinase activation was also dependent upon PDGFRβ and its tyrosine phosphorylation at 740/751 residues. Inhibition of PDGFRβ activity, its downregulation and expression of its phospho-deficient (Y740/751F) mutant inhibited mesangial cell hypertrophy by high glucose. Interestingly, expression of constitutively active Akt reversed this inhibition, indicating a role of Akt kinase downstream of PDGFRβ phosphorylation in this process. The transcription factor Hif1α is a target of Akt kinase. siRNAs against Hif1α inhibited the high glucose-induced mesangial cell hypertrophy. In contrast, increased expression of Hif1α induced hypertrophy similar to high glucose. We found that inhibition of PDGFRβ and expression of PDGFRβ Y740/751F mutant significantly inhibited the high glucose-induced expression of Hif1α. Importantly, expression of Hif1α countered the inhibition of mesangial cell hypertrophy induced by siPDGFRβ or PDGFRβ Y740/751F mutant. Finally, we show that high glucose-stimulated PDGFRβ tyrosine phosphorylation at 740/751 residues and the tyrosine kinase activity of the receptor regulate the transforming growth factor-β (TGFβ) expression by Hif1α. Thus we define the cell surface PDGFRβ as a major link between high glucose and its effectors Hif1α and TGFβ for induction of diabetic mesangial cell hypertrophy.
Collapse
Affiliation(s)
- Falguni Das
- Department of Medicine, UT Health at San Antonio, TX, United States
| | - Nandini Ghosh-Choudhury
- VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, TX, United States; Department of Pathology, UT Health at San Antonio, TX, United States
| | - Balakuntalam S Kasinath
- Department of Medicine, UT Health at San Antonio, TX, United States; VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, TX, United States
| | - Goutam Ghosh Choudhury
- Department of Medicine, UT Health at San Antonio, TX, United States; VA Biomedical Laboratory Research, South Texas Veterans Health Care System, San Antonio, TX, United States; Geriatric Research, Education and Clinical Research, South Texas Veterans Health Care System, San Antonio, TX, United States.
| |
Collapse
|
45
|
Zeng B, Huang Y, Xu J, Shiotsuki T, Bai H, Palli SR, Huang Y, Tan A. The FOXO transcription factor controls insect growth and development by regulating juvenile hormone degradation in the silkworm, Bombyx mori. J Biol Chem 2017; 292:11659-11669. [PMID: 28490635 DOI: 10.1074/jbc.m117.777797] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 05/08/2017] [Indexed: 01/09/2023] Open
Abstract
Forkhead box O (FOXO) functions as the terminal transcription factor of the insulin signaling pathway and regulates multiple physiological processes in many organisms, including lifespan in insects. However, how FOXO interacts with hormone signaling to modulate insect growth and development is largely unknown. Here, using the transgene-based CRISPR/Cas9 system, we generated and characterized mutants of the silkworm Bombyx mori FOXO (BmFOXO) to elucidate its physiological functions during development of this lepidopteran insect. The BmFOXO mutant (FOXO-M) exhibited growth delays from the first larval stage and showed precocious metamorphosis, pupating at the end of the fourth instar (trimolter) rather than at the end of the fifth instar as in the wild-type (WT) animals. However, different from previous reports on precocious metamorphosis caused by juvenile hormone (JH) deficiency in silkworm mutants, the total developmental time of the larval period in the FOXO-M was comparable with that of the WT. Exogenous application of 20-hydroxyecdysone (20E) or of the JH analog rescued the trimolter phenotype. RNA-seq and gene expression analyses indicated that genes involved in JH degradation but not in JH biosynthesis were up-regulated in the FOXO-M compared with the WT animals. Moreover, we identified several FOXO-binding sites in the promoter of genes coding for JH-degradation enzymes. These results suggest that FOXO regulates JH degradation rather than its biosynthesis, which further modulates hormone homeostasis to control growth and development in B. mori In conclusion, we have uncovered a pivotal role for FOXO in regulating JH signaling to control insect development.
Collapse
Affiliation(s)
- Baosheng Zeng
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuping Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jun Xu
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Takahiro Shiotsuki
- National Agriculture and Food Research Organization, Institute of Agrobiological Sciences, Division of Insect Science, 1-2 Owashi, Tsukuba, Ibaraki 305-8634, Japan
| | - Hua Bai
- Department of Genetics, Development, and Cell Biology, Iowa State University, Ames, Iowa 50011
| | - Subba Reddy Palli
- Department of Entomology, University of Kentucky, Lexington, Kentucky 40546-0091
| | - Yongping Huang
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Anjiang Tan
- Key Laboratory of Insect Developmental and Evolutionary Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China.
| |
Collapse
|
46
|
Wakabayashi S, Sawamura N, Voelzmann A, Broemer M, Asahi T, Hoch M. Ohgata, the Single Drosophila Ortholog of Human Cereblon, Regulates Insulin Signaling-dependent Organismic Growth. J Biol Chem 2016; 291:25120-25132. [PMID: 27702999 PMCID: PMC5122779 DOI: 10.1074/jbc.m116.757823] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Indexed: 11/06/2022] Open
Abstract
Cereblon (CRBN) is a substrate receptor of the E3 ubiquitin ligase complex that is highly conserved in animals and plants. CRBN proteins have been implicated in various biological processes such as development, metabolism, learning, and memory formation, and their impairment has been linked to autosomal recessive non-syndromic intellectual disability and cancer. Furthermore, human CRBN was identified as the primary target of thalidomide teratogenicity. Data on functional analysis of CRBN family members in vivo, however, are still scarce. Here we identify Ohgata (OHGT), the Drosophila ortholog of CRBN, as a regulator of insulin signaling-mediated growth. Using ohgt mutants that we generated by targeted mutagenesis, we show that its loss results in increased body weight and organ size without changes of the body proportions. We demonstrate that ohgt knockdown in the fat body, an organ analogous to mammalian liver and adipose tissue, phenocopies the growth phenotypes. We further show that overgrowth is due to an elevation of insulin signaling in ohgt mutants and to the down-regulation of inhibitory cofactors of circulating Drosophila insulin-like peptides (DILPs), named acid-labile subunit and imaginal morphogenesis protein-late 2. The two inhibitory proteins were previously shown to be components of a heterotrimeric complex with growth-promoting DILP2 and DILP5. Our study reveals OHGT as a novel regulator of insulin-dependent organismic growth in Drosophila.
Collapse
Affiliation(s)
- Satoru Wakabayashi
- From the Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Naoya Sawamura
- From the Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan,
- the Research Organization for Nano-life Innovation, Waseda University, Shinjuku, Tokyo 162-0041, Japan
| | - André Voelzmann
- the Faculty of Biology, Medicine and Health, The University of Manchester, Michael Smith Building, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Meike Broemer
- the German Center for Neurodegenerative Diseases (DZNE), c/o Life and Medical Sciences (LIMES) Institute, Carl-Troll-Strasse 31, 53115 Bonn, Germany, and
| | - Toru Asahi
- From the Faculty of Science and Engineering, Waseda University, TWIns, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan,
- the Research Organization for Nano-life Innovation, Waseda University, Shinjuku, Tokyo 162-0041, Japan
| | - Michael Hoch
- Program Unit Development, Genetics and Molecular Physiology, Laboratory for Molecular Developmental Biology, LIMES Institute, University of Bonn, Carl-Troll-Strasse 31, 53115 Bonn, Germany
| |
Collapse
|
47
|
Altintas O, Park S, Lee SJV. The role of insulin/IGF-1 signaling in the longevity of model invertebrates, C. elegans and D. melanogaster. BMB Rep 2016; 49:81-92. [PMID: 26698870 PMCID: PMC4915121 DOI: 10.5483/bmbrep.2016.49.2.261] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Indexed: 01/08/2023] Open
Abstract
Insulin/insulin-like growth factor (IGF)-1 signaling (IIS) pathway regulates
aging in many organisms, ranging from simple invertebrates to mammals, including
humans. Many seminal discoveries regarding the roles of IIS in aging and
longevity have been made by using the roundworm Caenorhabditis
elegans and the fruit fly Drosophila melanogaster. In this
review, we describe the mechanisms by which various IIS components regulate
aging in C. elegans and D. melanogaster. We
also cover systemic and tissue-specific effects of the IIS components on the
regulation of lifespan. We further discuss IIS-mediated physiological processes
other than aging and their effects on human disease models focusing on
C. elegans studies. As both C. elegans and
D. melanogaster have been essential for key findings
regarding the effects of IIS on organismal aging in general, these invertebrate
models will continue to serve as workhorses to help our understanding of
mammalian aging. [BMB Reports 2016; 49(2): 81-92]
Collapse
Affiliation(s)
- Ozlem Altintas
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Sangsoon Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Seung-Jae V Lee
- School of Interdisciplinary Bioscience and Bioengineering, Department of Life Sciences, and Information Technology Convergence Engineering, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
48
|
Murillo-Maldonado JM, Riesgo-Escovar JR. Development and diabetes on the fly. Mech Dev 2016; 144:150-155. [PMID: 27702607 DOI: 10.1016/j.mod.2016.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 09/30/2016] [Accepted: 09/30/2016] [Indexed: 10/20/2022]
Abstract
We review the use of a model organism to study the effects of a slow course, degenerative disease: namely, diabetes mellitus. Development and aging are biological phenomena entailing reproduction, growth, and differentiation, and then decline and progressive loss of functionality leading ultimately to failure and death. It occurs at all biological levels of organization, from molecular interactions to organismal well being and homeostasis. Yet very few models capable of addressing the different levels of complexity in these chronic, developmental phenomena are available to study, and model organisms are an exception and a welcome opportunity for these approaches. Genetic model organisms, like the common fruit fly, Drosophila melanogaster, offer the possibility of studying the panoply of life processes in normal and diseased states like diabetes mellitus, from a plethora of different perspectives. These long-term aspects are now beginning to be characterized.
Collapse
Affiliation(s)
- Juan Manuel Murillo-Maldonado
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Boulevard Juriquilla #3001, Querétaro 76230, Mexico
| | - Juan Rafael Riesgo-Escovar
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM Juriquilla, Boulevard Juriquilla #3001, Querétaro 76230, Mexico.
| |
Collapse
|
49
|
Cell Competition and Its Role in the Regulation of Cell Fitness from Development to Cancer. Dev Cell 2016; 38:621-34. [DOI: 10.1016/j.devcel.2016.08.012] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/01/2016] [Accepted: 08/22/2016] [Indexed: 12/26/2022]
|
50
|
Otero-Moreno D, Peña-Rangel MT, Riesgo-Escovar JR. CRECIMIENTO Y METABOLISMO: LA REGULACIÓN Y LA VÍA DE LA INSULINA DESDE LA MOSCA DE LA FRUTA, Drosophila melanogaster. TIP REVISTA ESPECIALIZADA EN CIENCIAS QUÍMICO-BIOLÓGICAS 2016. [DOI: 10.1016/j.recqb.2016.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|