1
|
Xiao G, Li Y, Hu Y, Tan K, Wang M, Zhu K, San M, Cheng Q, Tayier D, Hu T, Dang P, Li J, Cheng C, Perrimon N, Yang Z, Song W. Intratumor HIF-1α modulates production of a cachectic ligand to cause host wasting. CELL INSIGHT 2025; 4:100247. [PMID: 40336592 PMCID: PMC12056967 DOI: 10.1016/j.cellin.2025.100247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Tumor-host interactions play critical roles in cancer-associated cachexia. Previous studies have identified several cachectic proteins secreted by tumors that impair metabolic homeostasis in multiple organs, leading to host wasting. The molecular mechanisms by which malignant tumors regulate the production or secretion of these cachectic proteins, however, still remain largely unknown. In this study, we used different Drosophila cachexia models to investigate how malignant tumors regulate biosynthesis of ImpL2, a conserved cachectic protein that inhibits systemic insulin/IGF signaling and suppresses anabolism of host organs. Through bioinformatic and biochemical analysis, we found that hypoxia-inducible factor HIF-1α/Sima directly binds to the promoter region of ImpL2 gene for the first time, promoting its transcription in both tumors and non-tumor cells. Interestingly, expressing HphA to moderately suppress HIF-1α/Sima activity in adult yki 3SA gut tumors or larval scrib 1 Ras V12 disc tumors sufficiently decreased ImpL2 expression and improved organ wasting, without affecting tumor growth. We further revealed conserved regulatory mechanisms conserved across species, as intratumor HIF-1α enhances the production of IGFBP-5, a mammalian homolog of fly ImpL2, contributing to organ wasting in both tumor-bearing mice and patients. Therefore, our study provides novel insights into the mechanisms by which tumors regulate production of cachectic ligands and the pathogenesis of cancer-induced cachexia.
Collapse
Affiliation(s)
- Gen Xiao
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yingge Li
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Yanhui Hu
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Kai Tan
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
| | - Mengyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
| | - Kerui Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
| | - Mingkui San
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
| | - Qian Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
| | - Dilinigeer Tayier
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Tingting Hu
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Peixuan Dang
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Jiaying Li
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| | - Norbert Perrimon
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Zhiyong Yang
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, Hubei, China
| |
Collapse
|
2
|
Park JS, Sung MJ, Na HJ. Drosophila model systems reveal intestinal stem cells as key players in aging. Ann N Y Acad Sci 2025; 1547:88-99. [PMID: 40276941 DOI: 10.1111/nyas.15351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
The intestines play important roles in responding immediately and dynamically to food intake, environmental stress, and metabolic dysfunction, and they are involved in various human diseases and aging. A key part of their function is governed by intestinal stem cells (ISCs); therefore, understanding ISCs is vital. Dysregulation of ISC activity, which is influenced by various cell signaling pathways and environmental signals, can lead to inflammatory responses, tissue damage, and increased cancer susceptibility. Aging exacerbates these dynamics and affects ISC function and tissue elasticity. Additionally, proliferation and differentiation profoundly affect ISC behavior and gut health, highlighting the complex interplay between environmental factors and gut homeostasis. Drosophila models help us understand the complex regulatory networks in the gut, providing valuable insights into disease mechanisms and therapeutic strategies targeting human intestinal diseases.
Collapse
Affiliation(s)
- Joung-Sun Park
- Institute of Nanobio Convergence, Pusan National University, Busan, Republic of Korea
- Department of Molecular Biology, Pusan National University, Busan, Republic of Korea
| | - Mi Jeong Sung
- Aging Research Group, Division of Food Functionality Research, Korea Food Research Institute, Wanju, Republic of Korea
| | - Hyun-Jin Na
- Aging Research Group, Division of Food Functionality Research, Korea Food Research Institute, Wanju, Republic of Korea
| |
Collapse
|
3
|
Tsai M, Sun J, Alexandre C, Shapiro M, Franchet A, Li Y, Gould AP, Vincent JP, Stockinger B, Diny NL. Drosophila AHR limits tumor growth and stem cell proliferation in the intestine. Wellcome Open Res 2025; 10:38. [PMID: 40212817 PMCID: PMC11982807 DOI: 10.12688/wellcomeopenres.23515.2] [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] [Accepted: 04/23/2025] [Indexed: 04/29/2025] Open
Abstract
Background The aryl hydrocarbon receptor (AHR) plays important roles in intestinal homeostasis, limiting tumour growth and promoting differentiation in the intestinal epithelium. Spineless, the Drosophila homolog of AHR, has only been studied in the context of development but not in the adult intestine. Methods The role of Spineless in the Drosophila midgut was studied by overexpression or inactivation of Spineless in infection and tumour models and RNA sequencing of sorted midgut progenitor cells. Results We show that spineless is upregulated in the adult intestinal epithelium after infection with Pseudomonas entomophila ( P. e.). Spineless inactivation increased stem cell proliferation following infection-induced injury. Spineless overexpression limited intestinal stem cell proliferation and reduced survival after infection. In two tumour models, using either Notch RNAi or constitutively active Yorkie, Spineless suppressed tumour growth and doubled the lifespan of tumour-bearing flies. At the transcriptional level it reversed the gene expression changes induced in Yorkie tumours, counteracting cell proliferation and altered metabolism. Conclusions These findings demonstrate a new role for Spineless in the adult Drosophila midgut and highlight the evolutionarily conserved functions of AHR/Spineless in the control of proliferation and differentiation of the intestinal epithelium.
Collapse
Affiliation(s)
- Minghua Tsai
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Jiawei Sun
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | | | | | | | - Ying Li
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | - Alex P. Gould
- The Francis Crick Institute, London, England, NW1 1AT, UK
| | | | | | - Nicola Laura Diny
- The Francis Crick Institute, London, England, NW1 1AT, UK
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, North Rhine-Westphalia, 53127, Germany
| |
Collapse
|
4
|
Ma X, Li H, Li Y, Xie X, Wang Y, Wang M, Peng X. Potential Antidiabetic Activity of Nordihydroguaiaretic Acid: An Insight into Its Inhibitory Mechanisms on Carbohydrate-Hydrolyzing Enzymes, the Binding Behaviors with Enzymes, and In Vivo Antihyperglycemic Effect. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:8290-8304. [PMID: 40152424 DOI: 10.1021/acs.jafc.4c11307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
The inhibitory mechanisms and binding behaviors of nordihydroguaiaretic acid (NDGA) to α-glucosidase/α-amylase were investigated by in vitro multispectroscopic methods and in silico modeling technique. The results demonstrated that NDGA reversibly and uncompetitively inhibited α-glucosidase, exhibiting stronger inhibition than acarbose, while it displayed noncompetitive inhibition against α-amylase. Additionally, NDGA could spontaneously bind to α-glucosidase/α-amylase mainly through hydrogen bonds and hydrophobic forces, thus altering the spatial structure of enzymes and reducing their catalytic activity. The presence of crowding reagents/polysaccharides/undigested milk proteins would decrease the inhibitory ability of NDGA, whereas fatty acids exhibited the opposite phenomenon on α-glucosidase. Furthermore, the antidiabetic activity of NDGA in vivo was evaluated using the diabetic Drosophila model induced by a high-sugar diet. It was found that NDGA significantly reduced the glucose levels of diabetic Drosophila. These findings suggested that NDGA was a potential inhibitor of α-glucosidase/α-amylase and could be used as a nutritional adjuvant to prevent diabetes.
Collapse
Affiliation(s)
- Xiangzhao Ma
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Huan Li
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ying Li
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xiaofang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Mengfan Wang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Xin Peng
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
5
|
Huang K, Miao T, Dantas E, Han M, Hu Y, Wang K, Sanford J, Goncalves M, Perrimon N. Lipid metabolism of hepatocyte-like cells supports intestinal tumor growth by promoting tracheogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647255. [PMID: 40236168 PMCID: PMC11996582 DOI: 10.1101/2025.04.04.647255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Tumors require metabolic adaptations to support their rapid growth, but how they influence lipid metabolism in distant tissues remains poorly understood. Here, we uncover a novel mechanism by which gut tumors in adult flies reprogram lipid metabolism in distal hepatocyte-like cells, known as oenocytes, to promote tracheal development and tumor growth. We show that tumors secrete a PDGF/VEGF-like factor, Pvf1, that activates the TORC1-Hnf4 signaling pathway in oenocytes. This activation enhances the production of specific lipids, including very long-chain fatty acids and wax esters, that are required for tracheal growth surrounding the gut tumor. Importantly, reducing expression in oenocytes of either the transcription factor Hnf4 , or the elongase mElo that generates very long chain fatty acid suppresses tumor growth, tracheogenesis, and associated organ wasting/cachexia-like phenotypes, while extending lifespan. We further demonstrate that this regulatory pathway is conserved in mammals, as VEGF-A stimulates lipid metabolism gene expression in human hepatocytes, and lung tumor-bearing mice show increased hepatic expression of Hnf4 and the lipid elongation gene Elovl7 . Our findings reveal a previously unrecognized tumor-host interaction where tumors non-autonomously reprogram distal lipid metabolism to support their growth. This study not only identifies a novel non-autonomous role of the TORC1-Hnf4 axis in lipid-mediated tumor progression but also highlights potential targets for therapeutic intervention in cancer-associated metabolic disorders.
Collapse
|
6
|
Liu Y, Dantas E, Ferrer M, Miao T, Qadiri M, Liu Y, Comjean A, Davidson EE, Perrier T, Ahmed T, Hu Y, Goncalves MD, Janowitz T, Perrimon N. Hepatic gluconeogenesis and PDK3 upregulation drive cancer cachexia in flies and mice. Nat Metab 2025; 7:823-841. [PMID: 40275022 PMCID: PMC12021660 DOI: 10.1038/s42255-025-01265-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 03/06/2025] [Indexed: 04/26/2025]
Abstract
Cachexia, a severe wasting syndrome characterized by tumour-induced metabolic dysregulation, is a leading cause of death in people with cancer, yet its underlying mechanisms remain poorly understood. Here we show that a longitudinal full-body single-nuclei-resolution transcriptome analysis in a Drosophila model of cancer cachexia captures interorgan dysregulations. Our study reveals that the tumour-secreted interleukin-like cytokine Upd3 induces fat-body expression of Pepck1 and Pdk, key regulators of gluconeogenesis, disrupting glucose metabolism and contributing to cachexia. Similarly, in mouse cancer cachexia models, we observe IL-6-JAK-STAT-signalling-mediated induction of Pck1 and Pdk3 expression in the liver. Increased expression of these genes in fly, mouse, and human correlates with poor prognosis, and hepatic expression of Pdk3 emerges as a previously unknown mechanism contributing to metabolic dysfunction in cancer cachexia. This study highlights the conserved nature of tumour-induced metabolic disruptions and identifies potential therapeutic targets to mitigate cachexia in people with cancer.
Collapse
Affiliation(s)
- Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Ezequiel Dantas
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
| | - Ting Miao
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Mujeeb Qadiri
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Emma E Davidson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Ohio State University College of Medicine, Columbus, OH, USA
| | - Tiffany Perrier
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Tanvir Ahmed
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Marcus D Goncalves
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, USA
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, New York, NY, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
7
|
Aloisi M, Deloriea J, Casey C, Pino A, Morciano P, Grifoni D, Gamberi C. DROSOPHILA: THE CENTURY-LONG FLIGHT FROM THE WILD TO THE PATIENT. MEDICAL SCIENCE PULSE 2025; 19:1-15. [PMID: 40276781 PMCID: PMC12021435 DOI: 10.5604/01.3001.0054.9627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Abstract
Background: Evolutionary conservation of key biological pathways between the fruit fly Drosophila melanogaster and humans and reduced genetic redundancy have long made flies a valuable genetic model organism. Thanks to the arsenal of sophisticated genetic tools developed and refined by the fly community, the use of Drosophila has expanded well beyond basic research. From the fundamental notion that genes are located on chromosomes to modeling human complex diseases such as cancer and neurological disorders, to designing fly "avatar" lines that precisely reproduce the specific mutations found in single cancer patients for personalized medicine, Drosophila continues to fuel biomedical advances. Numerous examples of drug testing in flies have yielded novel drug candidates, new uses for approved drugs, and applications for rapid drug optimization in modern approaches combining biology with medicinal chemistry. Thanks to the effectiveness of "fly pharmacology" approaches, Drosophila is also proficiently used to study the mechanism of action of environmental pollutants that represent a serious concern to human health. This review traces the history of some of the main advances in Drosophila biomedical and cancer research.
Collapse
Affiliation(s)
- Massimo Aloisi
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
- Department of Life, Health and Environmental Sciences, L'Aquila University, Italy
| | - Jay Deloriea
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| | - Cody Casey
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| | - Alexia Pino
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| | - Patrizia Morciano
- Department of Life, Health and Environmental Sciences, L'Aquila University, Italy
- INFN-Laboratori Nazionali del Gran Sasso, Assergi, L'Aquila, Italy
| | - Daniela Grifoni
- Department of Life, Health and Environmental Sciences, L'Aquila University, Italy
| | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, South Carolina, USA
| |
Collapse
|
8
|
Rondeau NC, Raup-Collado J, Kogan HV, Cho R, Lovinger N, Wague F, Lopatkin AJ, Texeira NG, Flores ME, Rovnyak D, Snow JW. Remodeling of Cellular Respiration and Insulin Signaling Are Part of a Shared Stress Response in Divergent Bee Species. INSECTS 2025; 16:300. [PMID: 40266798 PMCID: PMC11942726 DOI: 10.3390/insects16030300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 04/25/2025]
Abstract
The honey bee (Apis mellifera) is of paramount importance to human activities through the pollination services they provide in agricultural settings. Honey bee colonies in the United States have suffered from an increased rate of annual die-off in recent years, stemming from a complex set of interacting stressors that remain poorly described. Defining the cellular responses that are perturbed by divergent stressors represents a key step in understanding these synergies. We found that multiple model stressors induce upregulated expression of the lactate dehydrogenase (Ldh) gene in the midgut of the eusocial honey bee and that the Ldh gene family is expanded in diverse bee species. Alterations in Ldh expression were concomitant with changes in the expression of other genes involved in cellular respiration and genes encoding insulin/insulin-like growth factor signaling (IIS) pathway components. Additionally, changes in metabolites in the midgut after stress, including increased levels of lactate, linked metabolic changes with the observed changes in gene expression. Select transcriptional changes in response to stress were similarly observed in the solitary alfalfa leafcutting bee (Megachile rotundata). Thus, increased Ldh expression may be part of a core stress response remodeling cellular respiration and insulin signaling. These findings suggest that a conserved cellular response that regulates metabolic demands under diverse stressful conditions may play a protective role in bees regardless of life history.
Collapse
Affiliation(s)
- Nicole C. Rondeau
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Joanna Raup-Collado
- Department of Chemistry, Bucknell University, Lewisburg, PA 17837, USA; (J.R.-C.); (D.R.)
| | - Helen V. Kogan
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Rachel Cho
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Natalie Lovinger
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Fatoumata Wague
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Allison J. Lopatkin
- Department of Chemical Engineering, University of Rochester, Rochester, NY 14642, USA;
| | - Noelle G. Texeira
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - Melissa E. Flores
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| | - David Rovnyak
- Department of Chemistry, Bucknell University, Lewisburg, PA 17837, USA; (J.R.-C.); (D.R.)
| | - Jonathan W. Snow
- Biology Department, Barnard College, New York, NY 10027, USA; (N.C.R.); (H.V.K.); (M.E.F.)
| |
Collapse
|
9
|
Hu Y, Rodiger J, Liu Y, Gao C, Liu Y, Qadiri M, Veal A, Bulyk ML, Perrimon N. TF2TG: an online resource mining the potential gene targets of transcription factors in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.638157. [PMID: 39990429 PMCID: PMC11844531 DOI: 10.1101/2025.02.13.638157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Sequence-specific transcription factors (TFs) are key regulators of many biological processes, controlling the expression of their target genes through binding to the cis- regulatory regions such as promoters and enhancers. Each TF has unique DNA binding site motifs, and large-scale experiments have been conducted to characterize TF-DNA binding preferences. However, no comprehensive resource currently integrates these datasets for Drosophila. To address this need, we developed TF2TG ("transcription factor to target gene"), a comprehensive resource that combines both in vitro and in vivo datasets to link transcription factors (TFs) to their target genes based on TF-DNA binding preferences along with the protein-protein interaction data, tissue-specific transcriptomic data, and chromatin accessibility data. Although the genome offers numerous potential binding sites for each TF, only a subset is actually bound in vivo, and of these, only a fraction is functionally relevant. For instance, some TFs bind to their specific sites due to synergistic interactions with other factors nearby. This integration provides users with a comprehensive list of potential candidates as well as aids users in ranking candidate genes and determining condition-specific TF binding for studying transcriptional regulation in Drosophila.
Collapse
Affiliation(s)
- Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Jonathan Rodiger
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Chenxi Gao
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Mujeeb Qadiri
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Austin Veal
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Martha L. Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Drosophila RNAi Screening Center, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02138, USA
| |
Collapse
|
10
|
Ding G, Li Y, Cheng C, Tan K, Deng Y, Pang H, Wang Z, Dang P, Wu X, Rushworth E, Yuan Y, Yang Z, Song W. A tumor-secreted protein utilizes glucagon release to cause host wasting. Cell Discov 2025; 11:11. [PMID: 39924534 PMCID: PMC11808122 DOI: 10.1038/s41421-024-00762-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 12/23/2024] [Indexed: 02/11/2025] Open
Abstract
Tumor‒host interaction plays a critical role in malignant tumor-induced organ wasting across multiple species. Despite known regulation of regional wasting of individual peripheral organs by tumors, whether and how tumors utilize critical host catabolic hormone(s) to simultaneously induce systemic host wasting, is largely unknown. Using the conserved yki3SA-tumor model in Drosophila, we discovered that tumors increase the production of adipokinetic hormone (Akh), a glucagon-like catabolic hormone, to cause systemic host wasting, including muscle dysfunction, lipid loss, hyperglycemia, and ovary atrophy. We next integrated RNAi screening and Gal4-LexA dual expression system to show that yki3SA-gut tumors secrete Pvf1 to remotely activate its receptor Pvr in Akh-producing cells (APCs), ultimately promoting Akh production. The underlying molecular mechanisms involved the Pvf1-Pvr axis that triggers Mmp2-dependent ECM remodeling of APCs and enhances innervation from the excitatory cholinergic neurons. Interestingly, we also confirmed the similar mechanisms governing tumor-induced glucagon release and organ wasting in mammals. Blockade of either glucagon or PDGFR (homolog of Pvr) action efficiently ameliorated organ wasting in the presence of malignant tumors. Therefore, our results demonstrate that tumors remotely promote neural-associated Akh/glucagon production via Pvf1-Pvr axis to cause systemic host wasting.
Collapse
Affiliation(s)
- Guangming Ding
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yingge Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Kai Tan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Yifei Deng
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Huiwen Pang
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Zhongyuan Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Peixuan Dang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xing Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Elisabeth Rushworth
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Zhiyong Yang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
11
|
Singh A, Hu Y, Lopes RF, Lane L, Woldemichael H, Xu C, Udeshi ND, Carr SA, Perrimon N. Cell-death induced immune response and coagulopathy promote cachexia in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631515. [PMID: 39829769 PMCID: PMC11741341 DOI: 10.1101/2025.01.07.631515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Tumors can exert a far-reaching influence on the body, triggering systemic responses that contribute to debilitating conditions like cancer cachexia. To characterize the mechanisms underlying tumor-host interactions, we utilized a BioID-based proximity labeling method to identify proteins secreted by Ykiact adult Drosophila gut tumors into the bloodstream/hemolymph. Among the major proteins identified are coagulation and immune-responsive factors that contribute to the systemic wasting phenotypes associated with Ykiact tumors. The effect of innate immunity factors is mediated by NFκB transcription factors Relish, dorsal, and Dif, which in turn upregulate the expression of the cachectic factors Pvf1, Impl2, and Upd3. In addition, Ykiact tumors secrete Eiger, a TNF-alpha homolog, which activates the JNK signaling pathway in neighboring non-tumor cells, leading to cell death. The release of damage-associated molecular patterns (DAMPs) from these dying cells presumably amplifies the inflammatory response, exacerbating systemic wasting. Targeting the inflammatory response, the JNK pathway, or the production of cachectic factors could potentially alleviate the debilitating effects of cancer cachexia.
Collapse
Affiliation(s)
- Ankita Singh
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | - Raphael Fragoso Lopes
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | - Liz Lane
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
| | | | - Charles Xu
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Steven A. Carr
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 7 02115, USA
- HHMI, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
12
|
Kumar R, Srikrishna S. JNK Kinase regulates cachexia like syndrome in scribble knockdown tumor model of Drosophila melanogaster. Dev Biol 2025; 517:28-38. [PMID: 39293747 DOI: 10.1016/j.ydbio.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 09/04/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Cachexia and systemic organ wasting are metabolic syndrome often associated with cancer. However, the exact mechanism of cancer associated cachexia like syndrome still remain elusive. In this study, we utilized a scribble (scrib) knockdown induced hindgut tumor to investigate the role of JNK kinase in cachexia like syndrome. Scrib, a cell polarity regulator, also acts as a tumor suppressor gene. Its loss and mis-localization are reported in various type of malignant cancer-like breast, colon and prostate cancer. The scrib knockdown flies exhibited male lethality, reduced life span, systemic organ wasting and increased pJNK level in hindgut of female flies. Interestingly, knocking down of human JNK Kinase analogue, hep, in scrib knockdown background in hindgut leads to restoration of loss of scrib mediated lethality and systemic organ wasting. Our data showed that scrib loss in hindgut is capable of inducing cancer associated cachexia like syndrome. Here, we firstly report that blocking the JNK signaling pathway effectively rescued the cancer cachexia induced by scrib knockdown, along with its associated gut barrier disruption. These findings have significantly advanced our understanding of cancer cachexia and have potential implications for the development of therapeutic strategies. However, more research is needed to fully understand the complex mechanisms underlying this condition.
Collapse
|
13
|
Berriel Diaz M, Rohm M, Herzig S. Cancer cachexia: multilevel metabolic dysfunction. Nat Metab 2024; 6:2222-2245. [PMID: 39578650 DOI: 10.1038/s42255-024-01167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 10/16/2024] [Indexed: 11/24/2024]
Abstract
Cancer cachexia is a complex metabolic disorder marked by unintentional body weight loss or 'wasting' of body mass, driven by multiple aetiological factors operating at various levels. It is associated with many malignancies and significantly contributes to cancer-related morbidity and mortality. With emerging recognition of cancer as a systemic disease, there is increasing awareness that understanding and treatment of cancer cachexia may represent a crucial cornerstone for improved management of cancer. Here, we describe the metabolic changes contributing to body wasting in cachexia and explain how the entangled action of both tumour-derived and host-amplified processes induces these metabolic changes. We discuss energy homeostasis and possible ways that the presence of a tumour interferes with or hijacks physiological energy conservation pathways. In that context, we highlight the role played by metabolic cross-talk mechanisms in cachexia pathogenesis. Lastly, we elaborate on the challenges and opportunities in the treatment of this devastating paraneoplastic phenomenon that arise from the complex and multifaceted metabolic cross-talk mechanisms and provide a status on current and emerging therapeutic approaches.
Collapse
Affiliation(s)
- Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany.
- Joint Heidelberg-IDC Translational Diabetes Program, Department of Inner Medicine, Heidelberg University Hospital, Heidelberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany.
| |
Collapse
|
14
|
Lawson ME, Jones GM, Runion M, Sims D, Young A, Briggs O, Long LJ, Tin Chi Chak S, Bose I, Rele CP, Reed LK. Gene model for the ortholog of ImpL2 in Drosophila simulans. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.000715. [PMID: 39677518 PMCID: PMC11645545 DOI: 10.17912/micropub.biology.000715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 11/18/2024] [Accepted: 11/28/2024] [Indexed: 12/17/2024]
Abstract
Gene model for the ortholog of Ecdysone-inducible gene L2 ( ImpL2 ) in the May 2017 (Princeton ASM75419v2/DsimGB2) Genome Assembly (GenBank Accession: GCA_000754195.3 ) of Drosophila simulans . This ortholog was characterized as part of a developing dataset to study the evolution of the Insulin/insulin-like growth factor signaling pathway (IIS) across the genus Drosophila using the Genomics Education Partnership gene annotation protocol for Course-based Undergraduate Research Experiences.
Collapse
Affiliation(s)
| | | | | | - Drew Sims
- Oklahoma Christian University, Edmond, OK USA
| | | | | | | | | | | | | | - Laura K Reed
- Biological Sciences, The University of Alabama, Tuscaloosa, AL 35487
| |
Collapse
|
15
|
Saez-Carrion E, Aguilar-Aragon M, García-López L, Dominguez M, Uribe ML. Metabolic Adaptations in Cancer and the Host Using Drosophila Models and Advanced Tools. Cells 2024; 13:1977. [PMID: 39682725 PMCID: PMC11640731 DOI: 10.3390/cells13231977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer is a multifactorial process involving genetic, epigenetic, physiological, and metabolic changes. The ability of tumours to regulate new reactive pathways is essential for their survival. A key aspect of this involves the decision-making process of cancer cells as they balance the exploitation of surrounding and distant tissues for their own benefit while avoiding the rapid destruction of the host. Nutrition plays a central role in these processes but is inherently limited. Understanding how tumour cells interact with non-tumoural tissues to acquire nutrients is crucial. In this review, we emphasise the utility of Drosophila melanogaster as a model organism for dissecting the complex oncogenic networks underlying these interactions. By studying various levels-from individual tumour cells to systemic markers-we can gain new insights into how cancer adapts and thrives. Moreover, developing innovative technologies, such as high-throughput methods and metabolic interventions, enhances our ability to explore how tumours adapt to different conditions. These technological advances allow us to explore tumour adaptations and open new opportunities for potential therapeutic strategies.
Collapse
Affiliation(s)
- Ernesto Saez-Carrion
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Miguel Hernández (UMH), Campus de Sant Joan, 03550 Sant Joan d’Alacant, Spain; (M.A.-A.); (L.G.-L.); (M.D.)
| | - Mario Aguilar-Aragon
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Miguel Hernández (UMH), Campus de Sant Joan, 03550 Sant Joan d’Alacant, Spain; (M.A.-A.); (L.G.-L.); (M.D.)
| | - Lucia García-López
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Miguel Hernández (UMH), Campus de Sant Joan, 03550 Sant Joan d’Alacant, Spain; (M.A.-A.); (L.G.-L.); (M.D.)
- Faculty of Health Sciences, Universidad Europea de Valencia, 03016 Alicante, Spain
| | - Maria Dominguez
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Miguel Hernández (UMH), Campus de Sant Joan, 03550 Sant Joan d’Alacant, Spain; (M.A.-A.); (L.G.-L.); (M.D.)
| | - Mary Luz Uribe
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC), Universidad Miguel Hernández (UMH), Campus de Sant Joan, 03550 Sant Joan d’Alacant, Spain; (M.A.-A.); (L.G.-L.); (M.D.)
| |
Collapse
|
16
|
Zambrano-Tipan D, Narváez-Padilla V, Reynaud E. Escargot a Snail superfamily member and its multiple roles in Drosophila melanogaster development. J Cell Physiol 2024; 239:e31269. [PMID: 38572978 DOI: 10.1002/jcp.31269] [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: 12/01/2023] [Revised: 03/21/2024] [Accepted: 03/27/2024] [Indexed: 04/05/2024]
Abstract
The Snail superfamily of transcription factors plays a crucial role in metazoan development; one of the most important vertebrate members of this family is Snai1 which is orthologous to the Drosophila melanogaster esg gene. This review offers a comprehensive examination of the roles of the esg gene in Drosophila development, covering its expression pattern and downstream targets, and draws parallels between the vertebrate Snai1 family proteins on controlling the epithelial-to-mesenchymal transition and esg. This gene regulates stemness, ploidy, and pluripontency. esg is expressed in various tissues during development, including the gut, imaginal discs, and neuroblasts. The functions of the esg include the suppression of differentiation in intestinal stem cells and the preservation of diploidy in imaginal cells. In the nervous system development, esg expression also inhibits neuroblast differentiation, thus regulating the number of neurons and the moment in development of neuronal differentiation. Loss of esg function results in diverse developmental defects, including defects in intestinal stem cell maintenance and differentiation, and alters imaginal disc and nervous system development. Expression levels of esg also play a role in regulating longevity and metabolism in adult stages. This review provides an overview of the current understanding of esg's developmental role, emphasizing cellular and tissue effects that arise from its loss of function. The insights gained may contribute to a better understanding of evolutionary conserved developmental mechanisms and certain metabolic diseases.
Collapse
Affiliation(s)
- Diego Zambrano-Tipan
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| | - Verónica Narváez-Padilla
- Centro de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, México
| | - Enrique Reynaud
- Departamento de Genética del Desarrollo y Fisiología Molecular, Instituto de Biotecnología Universidad Nacional Autónoma de México, Cuernavaca, Morelos, México
| |
Collapse
|
17
|
Li L, Wazir J, Huang Z, Wang Y, Wang H. A comprehensive review of animal models for cancer cachexia: Implications for translational research. Genes Dis 2024; 11:101080. [PMID: 39220755 PMCID: PMC11364047 DOI: 10.1016/j.gendis.2023.101080] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 06/14/2023] [Accepted: 07/24/2023] [Indexed: 09/04/2024] Open
Abstract
Cancer cachexia is a multifactorial syndrome characterized by progressive weight loss and a disease process that nutritional support cannot reverse. Although progress has been made in preclinical research, there is still a long way to go in translating research findings into clinical practice. One of the main reasons for this is that existing preclinical models do not fully replicate the conditions seen in clinical patients. Therefore, it is important to understand the characteristics of existing preclinical models of cancer cachexia and pay close attention to the latest developments in preclinical models. The main models of cancer cachexia used in current research are allogeneic and xenograft models, genetically engineered mouse models, chemotherapy drug-induced models, Chinese medicine spleen deficiency models, zebrafish and Drosophila models, and cellular models. This review aims to revisit and summarize the commonly used animal models of cancer cachexia by evaluating existing preclinical models, to provide tools and support for translational medicine research.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Junaid Wazir
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Zhiqiang Huang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Yong Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| | - Hongwei Wang
- State Key Laboratory of Analytical Chemistry for Life Science, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, Jiangsu 210093, China
| |
Collapse
|
18
|
Kwok SH, Liu Y, Bilder D, Kim J. Paraneoplastic renal dysfunction in fly cancer models driven by inflammatory activation of stem cells. Proc Natl Acad Sci U S A 2024; 121:e2405860121. [PMID: 39392665 PMCID: PMC11494367 DOI: 10.1073/pnas.2405860121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/21/2024] [Indexed: 10/12/2024] Open
Abstract
Tumors can induce systemic disturbances in distant organs, leading to physiological changes that enhance host morbidity. In Drosophila cancer models, tumors have been known for decades to cause hypervolemic "bloating" of the abdominal cavity. Here we use allograft and transgenic tumors to show that hosts display fluid retention associated with autonomously defective secretory capacity of fly renal tubules, which function analogous to those of the human kidney. Excretion from these organs is blocked by abnormal cells that originate from inappropriate activation of normally quiescent renal stem cells (RSCs). Blockage is initiated by IL-6-like oncokines that perturb renal water-transporting cells and trigger a damage response in RSCs that proceeds pathologically. Thus, a chronic inflammatory state produced by the tumor causes paraneoplastic fluid dysregulation by altering cellular homeostasis of host renal units.
Collapse
Affiliation(s)
- Sze Hang Kwok
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Yuejiang Liu
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA94720
| | - David Bilder
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley, CA94720
| | - Jung Kim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, China
| |
Collapse
|
19
|
Rodríguez-Vázquez M, Falconi J, Heron-Milhavet L, Lassus P, Géminard C, Djiane A. Fat body glycolysis defects inhibit mTOR and promote distant muscle disorganization through TNF-α/egr and ImpL2 signaling in Drosophila larvae. EMBO Rep 2024; 25:4410-4432. [PMID: 39251827 PMCID: PMC11467327 DOI: 10.1038/s44319-024-00241-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 07/29/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
The fat body in Drosophila larvae functions as a reserve tissue and participates in the regulation of organismal growth and homeostasis through its endocrine activity. To better understand its role in growth coordination, we induced fat body atrophy by knocking down several key enzymes of the glycolytic pathway in adipose cells. Our results show that impairing the last steps of glycolysis leads to a drastic drop in adipose cell size and lipid droplet content, and downregulation of the mTOR pathway and REPTOR transcriptional activity. Strikingly, fat body atrophy results in the distant disorganization of body wall muscles and the release of muscle-specific proteins in the hemolymph. Furthermore, we showed that REPTOR activity is required for fat body atrophy downstream of glycolysis inhibition, and that the effect of fat body atrophy on muscles depends on the production of TNF-α/egr and of the insulin pathway inhibitor ImpL2.
Collapse
Affiliation(s)
| | | | | | - Patrice Lassus
- IRCM, Univ Montpellier, Inserm, ICM, CNRS, Montpellier, France
| | | | | |
Collapse
|
20
|
Bhattacharya R, Kumari J, Banerjee S, Tripathi J, Parihar SS, Mohan N, Sinha P. Hippo effector, Yorkie, is a tumor suppressor in select Drosophila squamous epithelia. Proc Natl Acad Sci U S A 2024; 121:e2319666121. [PMID: 39288176 PMCID: PMC11441523 DOI: 10.1073/pnas.2319666121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Mammalian Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) and Drosophila Yorkie (Yki) are transcription cofactors of the highly conserved Hippo signaling pathway. It has been long assumed that the YAP/TAZ/Yki signaling drives cell proliferation during organ growth. However, its instructive role in regulating developmentally programmed organ growth, if any, remains elusive. Out-of-context gain of YAP/TAZ/Yki signaling often turns oncogenic. Paradoxically, mechanically strained, and differentiated squamous epithelia display developmentally programmed constitutive nuclear YAP/TAZ/Yki signaling. The unknown, therefore, is how a growth-promoting YAP/TAZ/Yki signaling restricts proliferation in differentiated squamous epithelia. Here, we show that reminiscent of a tumor suppressor, Yki negatively regulates the cell growth-promoting PI3K/Akt/TOR signaling in the squamous epithelia of Drosophila tubular organs. Thus, downregulation of Yki signaling in the squamous epithelium of the adult male accessory gland (MAG) up-regulates PI3K/Akt/TOR signaling, inducing cell hypertrophy, exit from their cell cycle arrest, and, finally, culminating in squamous cell carcinoma (SCC). Thus, blocking PI3K/Akt/TOR signaling arrests Yki loss-induced MAG-SCC. Further, MAG-SCCs, like other lethal carcinomas, secrete a cachectin, Impl2-the Drosophila homolog of mammalian IGFBP7-inducing cachexia and shortening the lifespan of adult males. Moreover, in the squamous epithelium of other tubular organs, like the dorsal trunk of larval tracheal airways or adult Malpighian tubules, downregulation of Yki signaling triggers PI3K/Akt/TOR-induced cell hypertrophy. Our results reveal that Yki signaling plays an instructive, antiproliferative role in the squamous epithelia of tubular organs.
Collapse
Affiliation(s)
- Rachita Bhattacharya
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Jaya Kumari
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Shweta Banerjee
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Jyoti Tripathi
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Saurabh Singh Parihar
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Nitin Mohan
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| | - Pradip Sinha
- Biological Sciences and Bioengineering, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
- Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Uttar Pradesh208016, India
| |
Collapse
|
21
|
Teles-Reis J, Jain A, Liu D, Khezri R, Micheli S, Gomez AA, Dillard C, Rusten TE. EyaHOST, a modular genetic system for investigation of intercellular and tumor-host interactions in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.06.611647. [PMID: 39314415 PMCID: PMC11418954 DOI: 10.1101/2024.09.06.611647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Cell biology and genetic analysis of intracellular, intercellular and inter-organ interaction studies in animal models are key for understanding development, physiology, and disease. The MARCM technique can emulate tumor development by simultaneous clonal tumor suppressor loss-of-function generation coupled with GAL4-UAS-driven oncogene and marker expression, but the utility is limited for studying tumor-host interactions due to genetic constraints. To overcome this, we introduce EyaHOST, a novel system that replaces MARCM with the QF2-QUAS binary gene expression system under the eya promoter control, unleashing the fly community genome-wide GAL4-UAS driven tools to manipulate any host cells or tissue at scale. EyaHOST generates epithelial clones in the eye epithelium similar to MARCM. EyaHOST-driven Ras V12 oncogene overexpression coupled with scribble tumor suppressor knockdown recapitulates key cancer features, including systemic catabolic switching and organ wasting. We demonstrate effective tissue-specific manipulation of host compartments such as neighbouring epithelial cells, immune cells, fat body, and muscle using fly avatars with tissue-specific GAL4 drivers. Organ-specific inhibition of autophagy or stimulation of growth-signaling through PTEN knockdown in fat body or muscle prevents cachexia-like wasting. Additionally, we show that Ras V12 , scrib RNAi tumors induce caspase-driven apoptosis in the epithelial microenvironment. Inhibition of apoptosis by p35 expression in the microenvironment promotes tumor growth. EyaHOST offers a versatile modular platform for dissecting tumor-host interactions and other mechanisms involving intercellular and inter-organ communication in Drosophila . Highlights * eyes absent , eye disc-specific enhancer drives clonal KD recombinase flip-out activated QF2 expression in the larval eye epithelium for simultaneous QUAS-driven gain and loss-of-function analysis of gene function. *Clones are visualized by QUAS-tagBFP or QUAS-eGFP facilitating analysis of existing fluorescent reporters.*The GAL4-UAS system and existing genome-wide genetic tools are released to independently manipulate any cell population in the animal for cell biology, intercellular or inter-organ analysis for developmental, physiological, or disease model analysis.*Fly avatars for tumor-host interaction studies with multiple organs allow live monitoring and manipulation of tumors and organs in translucent larva.
Collapse
|
22
|
Chang CW, Chin YH, Liu MS, Shen YC, Yan SJ. High sugar diet promotes tumor progression paradoxically through aberrant upregulation of pepck1. Cell Mol Life Sci 2024; 81:396. [PMID: 39261338 PMCID: PMC11390995 DOI: 10.1007/s00018-024-05438-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/16/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
High dietary sugar (HDS), a contemporary dietary concern due to excessive intake of added sugars and carbohydrates, escalates the risk of metabolic disorders and concomitant cancers. However, the molecular mechanisms underlying HDS-induced cancer progression are not completely understood. We found that phosphoenolpyruvate carboxykinase 1 (PEPCK1), a pivotal enzyme in gluconeogenesis, is paradoxically upregulated in tumors by HDS, but not by normal dietary sugar (NDS), during tumor progression. Targeted knockdown of pepck1, but not pepck2, specifically in tumor tissue in Drosophila in vivo, not only attenuates HDS-induced tumor growth but also significantly improves the survival of Ras/Src tumor-bearing animals fed HDS. Interestingly, HP1a-mediated heterochromatin interacts directly with the pepck1 gene and downregulates pepck1 gene expression in wild-type Drosophila. Mechanistically, we demonstrated that, under HDS conditions, pepck1 knockdown reduces both wingless and TOR signaling, decreases evasion of apoptosis, reduces genome instability, and suppresses glucose uptake and trehalose levels in tumor cells in vivo. Moreover, rational pharmacological inhibition of PEPCK1, using hydrazinium sulfate, greatly improves the survival of tumor-bearing animals with pepck1 knockdown under HDS. This study is the first to show that elevated levels of dietary sugar induce aberrant upregulation of PEPCK1, which promotes tumor progression through altered cell signaling, evasion of apoptosis, genome instability, and reprogramming of carbohydrate metabolism. These findings contribute to our understanding of the complex relationship between diet and cancer at the molecular, cellular, and organismal levels and reveal PEPCK1 as a potential target for the prevention and treatment of cancers associated with metabolic disorders.
Collapse
Affiliation(s)
- Che-Wei Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, Taiwan
- Department of Physiology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, Taiwan
| | - Yu-Hshun Chin
- Department of Physiology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, Taiwan
| | - Meng-Syuan Liu
- Department of Physiology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, Taiwan
| | - Yu-Chia Shen
- Department of Physiology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, Taiwan
| | - Shian-Jang Yan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, Taiwan.
- Department of Physiology, College of Medicine, National Cheng Kung University, No. 1, University Road, Tainan City, Taiwan.
| |
Collapse
|
23
|
Pranoto IKA, Kwon YV. Protocol to analyze Drosophila intestinal tumor cellular heterogeneity using immunofluorescence imaging and nuclear size quantification. STAR Protoc 2024; 5:102946. [PMID: 38470911 PMCID: PMC10945268 DOI: 10.1016/j.xpro.2024.102946] [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: 12/07/2023] [Revised: 01/22/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Drosophila intestinal tumors show an extended cellular heterogeneity. We devise a protocol to assess tumor cell heterogeneity by employing nuclear size measurement and immunofluorescence-based cell lineage analysis. We describe steps for intestinal dissection, staining, and imaging, followed by detailed procedures for nuclear size analysis. This approach detects overall heterogeneity across the entire tumor cell population and deviations within specific cell populations. The procedure is also applicable for analyzing the heterogeneity of wild-type intestinal cells in various contexts. For complete details on the use and execution of this protocol, please refer to Pranoto et al.1.
Collapse
Affiliation(s)
| | - Young V Kwon
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
24
|
Leung NY, Xu C, Li JSS, Ganguly A, Meyerhof GT, Regimbald-Dumas Y, Lane EA, Breault DT, He X, Perrimon N, Montell C. Gut tumors in flies alter the taste valence of an anti-tumorigenic bitter compound. Curr Biol 2024; 34:2623-2632.e5. [PMID: 38823383 PMCID: PMC11308992 DOI: 10.1016/j.cub.2024.04.082] [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: 08/25/2022] [Revised: 03/28/2024] [Accepted: 04/30/2024] [Indexed: 06/03/2024]
Abstract
The sense of taste is essential for survival, as it allows animals to distinguish between foods that are nutritious from those that are toxic. However, innate responses to different tastants can be modulated or even reversed under pathological conditions. Here, we examined whether and how the internal status of an animal impacts taste valence by using Drosophila models of hyperproliferation in the gut. In all three models where we expressed proliferation-inducing transgenes in intestinal stem cells (ISCs), hyperproliferation of ISCs caused a tumor-like phenotype in the gut. While tumor-bearing flies had no deficiency in overall food intake, strikingly, they exhibited an increased gustatory preference for aristolochic acid (ARI), which is a bitter and normally aversive plant-derived chemical. ARI had anti-tumor effects in all three of our gut hyperproliferation models. For other aversive chemicals we tested that are bitter but do not have anti-tumor effects, gut tumors did not affect avoidance behaviors. We demonstrated that bitter-sensing gustatory receptor neurons (GRNs) in tumor-bearing flies respond normally to ARI. Therefore, the internal pathology of gut hyperproliferation affects neural circuits that determine taste valence postsynaptic to GRNs rather than altering taste identity by GRNs. Overall, our data suggest that increased consumption of ARI may represent an attempt at self-medication. Finally, although ARI's potential use as a chemotherapeutic agent is limited by its known toxicity in the liver and kidney, our findings suggest that tumor-bearing flies might be a useful animal model to screen for novel anti-tumor drugs.
Collapse
Affiliation(s)
- Nicole Y Leung
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Chiwei Xu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Joshua Shing Shun Li
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Anindya Ganguly
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Geoff T Meyerhof
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA
| | - Yannik Regimbald-Dumas
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Elizabeth A Lane
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Xi He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | - Craig Montell
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA; Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
25
|
Fejzo MS. Hyperemesis gravidarum theories dispelled by recent research: a paradigm change for better care and outcomes. Trends Mol Med 2024; 30:530-540. [PMID: 38782680 DOI: 10.1016/j.molmed.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/27/2024] [Accepted: 04/10/2024] [Indexed: 05/25/2024]
Abstract
Nausea and vomiting (NVP) affect most pregnant women. At the severe end of the clinical spectrum, hyperemesis gravidarum (HG) can be life-threatening. The condition is fraught with misconceptions that have slowed progress and left women undertreated. Herein, recent scientific advances are presented that dispel common myths associated with HG related to maternal/offspring outcomes, etiology, and evolution. There is now strong evidence that (i) HG is associated with poor outcomes, (ii) a common cause of NVP and HG has been identified, and (iii) NVP is likely a protective evolutionary mechanism that occurs throughout the animal kingdom but is no longer necessary for human survival. Therefore, it is encouraging that we are finally on the cusp of testing treatments that may put an end to unnecessary suffering.
Collapse
Affiliation(s)
- Marlena Schoenberg Fejzo
- Center for Genetic Epidemiology, Department of Population and Public Health Sciences, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA.
| |
Collapse
|
26
|
Moon SJ, Hu Y, Dzieciatkowska M, Kim AR, Chen PL, Asara JM, D’Alessandro A, Perrimon N. Identification of high sugar diet-induced dysregulated metabolic pathways in muscle using tissue-specific metabolic models in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.24.591006. [PMID: 38712132 PMCID: PMC11071505 DOI: 10.1101/2024.04.24.591006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Individual tissues perform highly specialized metabolic functions to maintain whole-body homeostasis. Although Drosophila serves as a powerful model for studying human metabolic diseases, a lack of tissue-specific metabolic models makes it challenging to quantitatively assess the metabolic processes of individual tissues and disease models in this organism. To address this issue, we reconstructed 32 tissue-specific genome-scale metabolic models (GEMs) using pseudo-bulk single cell transcriptomics data, revealing distinct metabolic network structures across tissues. Leveraging enzyme kinetics and flux analyses, we predicted tissue-dependent metabolic pathway activities, recapitulating known tissue functions and identifying tissue-specific metabolic signatures, as supported by metabolite profiling. Moreover, to demonstrate the utility of tissue-specific GEMs in a disease context, we examined the effect of a high sugar diet (HSD) on muscle metabolism. Together with 13C-glucose isotopic tracer studies, we identified glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as a rate-limiting enzyme in response to HSD. Mechanistically, the decreased GAPDH activity was linked to elevated NADH/NAD+ ratio, caused by disturbed NAD+ regeneration rates, and oxidation of GAPDH. Furthermore, we introduced a pathway flux index to predict and validate additionally perturbed pathways, including fructose and butanoate metabolism. Altogether, our results represent a significant advance in generating quantitative tissue-specific GEMs and flux analyses in Drosophila, highlighting their use for identifying dysregulated metabolic pathways and their regulation in a human disease model.
Collapse
Affiliation(s)
- Sun Jin Moon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045
| | - Ah-Ram Kim
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - Po-Lin Chen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
| | - John M. Asara
- Division of Signal Transduction, Beth Israel Deaconess Medical Center, Boston, MA 02115
- Department of Medicine, Harvard Medical School, Boston, MA 02115
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado School of Medicine, Aurora, CO 80045
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
27
|
Kurogi Y, Mizuno Y, Kamiyama T, Niwa R. The intestinal stem cell/enteroblast-GAL4 driver, escargot-GAL4, also manipulates gene expression in the juvenile hormone-synthesizing organ of Drosophila melanogaster. Sci Rep 2024; 14:9631. [PMID: 38671036 PMCID: PMC11053112 DOI: 10.1038/s41598-024-60269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/21/2024] [Indexed: 04/28/2024] Open
Abstract
Intestinal stem cells (ISCs) of the fruit fly, Drosophila melanogaster, offer an excellent genetic model to explore homeostatic roles of ISCs in animal physiology. Among available genetic tools, the escargot (esg)-GAL4 driver, expressing the yeast transcription factor gene, GAL4, under control of the esg gene promoter, has contributed significantly to ISC studies. This driver facilitates activation of genes of interest in proximity to a GAL4-binding element, Upstream Activating Sequence, in ISCs and progenitor enteroblasts (EBs). While esg-GAL4 has been considered an ISC/EB-specific driver, recent studies have shown that esg-GAL4 is also active in other tissues, such as neurons and ovaries. Therefore, the ISC/EB specificity of esg-GAL4 is questionable. In this study, we reveal esg-GAL4 expression in the corpus allatum (CA), responsible for juvenile hormone (JH) production. When driving the oncogenic gene, RasV12, esg-GAL4 induces overgrowth in ISCs/EBs as reported, but also increases CA cell number and size. Consistent with this observation, animals alter expression of JH-response genes. Our data show that esg-GAL4-driven gene manipulation can systemically influence JH-mediated animal physiology, arguing for cautious use of esg-GAL4 as a "specific" ISC/EB driver to examine ISC/EB-mediated animal physiology.
Collapse
Affiliation(s)
- Yoshitomo Kurogi
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Yosuke Mizuno
- Graduate School of Science and Technology, University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Takumi Kamiyama
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8577, Japan
| | - Ryusuke Niwa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Ibaraki, 305-8577, Japan.
| |
Collapse
|
28
|
Ruan ZR, Yu Z, Xing C, Chen EH. Inter-organ steroid hormone signaling promotes myoblast fusion via direct transcriptional regulation of a single key effector gene. Curr Biol 2024; 34:1438-1452.e6. [PMID: 38513654 PMCID: PMC11003854 DOI: 10.1016/j.cub.2024.02.056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 12/24/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Steroid hormones regulate tissue development and physiology by modulating the transcription of a broad spectrum of genes. In insects, the principal steroid hormones, ecdysteroids, trigger the expression of thousands of genes through a cascade of transcription factors (TFs) to coordinate developmental transitions such as larval molting and metamorphosis. However, whether ecdysteroid signaling can bypass transcriptional hierarchies to exert its function in individual developmental processes is unclear. Here, we report that a single non-TF effector gene mediates the transcriptional output of ecdysteroid signaling in Drosophila myoblast fusion, a critical step in muscle development and differentiation. Specifically, we show that the 20-hydroxyecdysone (commonly referred to as "ecdysone") secreted from an extraembryonic tissue, amnioserosa, acts on embryonic muscle cells to directly activate the expression of antisocial (ants), which encodes an essential scaffold protein enriched at the fusogenic synapse. Not only is ants transcription directly regulated by the heterodimeric ecdysone receptor complex composed of ecdysone receptor (EcR) and ultraspiracle (USP) via ecdysone-response elements but also more strikingly, expression of ants alone is sufficient to rescue the myoblast fusion defect in ecdysone signaling-deficient mutants. We further show that EcR/USP and a muscle-specific TF Twist synergistically activate ants expression in vitro and in vivo. Taken together, our study provides the first example of a steroid hormone directly activating the expression of a single key non-TF effector gene to regulate a developmental process via inter-organ signaling and provides a new paradigm for understanding steroid hormone signaling in other developmental and physiological processes.
Collapse
Affiliation(s)
- Zhi-Rong Ruan
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ze Yu
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chao Xing
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabeth H Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
29
|
Dark C, Ali N, Golenkina S, Dhyani V, Blazev R, Parker BL, Murphy KT, Lynch GS, Senapati T, Millard SS, Judge SM, Judge AR, Giri L, Russell SM, Cheng LY. Mitochondrial fusion and altered beta-oxidation drive muscle wasting in a Drosophila cachexia model. EMBO Rep 2024; 25:1835-1858. [PMID: 38429578 PMCID: PMC11014992 DOI: 10.1038/s44319-024-00102-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/28/2024] [Accepted: 02/08/2024] [Indexed: 03/03/2024] Open
Abstract
Cancer cachexia is a tumour-induced wasting syndrome, characterised by extreme loss of skeletal muscle. Defective mitochondria can contribute to muscle wasting; however, the underlying mechanisms remain unclear. Using a Drosophila larval model of cancer cachexia, we observed enlarged and dysfunctional muscle mitochondria. Morphological changes were accompanied by upregulation of beta-oxidation proteins and depletion of muscle glycogen and lipid stores. Muscle lipid stores were also decreased in Colon-26 adenocarcinoma mouse muscle samples, and expression of the beta-oxidation gene CPT1A was negatively associated with muscle quality in cachectic patients. Mechanistically, mitochondrial defects result from reduced muscle insulin signalling, downstream of tumour-secreted insulin growth factor binding protein (IGFBP) homologue ImpL2. Strikingly, muscle-specific inhibition of Forkhead box O (FOXO), mitochondrial fusion, or beta-oxidation in tumour-bearing animals preserved muscle integrity. Finally, dietary supplementation with nicotinamide or lipids, improved muscle health in tumour-bearing animals. Overall, our work demonstrates that muscle FOXO, mitochondria dynamics/beta-oxidation and lipid utilisation are key regulators of muscle wasting in cancer cachexia.
Collapse
Affiliation(s)
- Callum Dark
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Nashia Ali
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Sofya Golenkina
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Vaibhav Dhyani
- Bioimaging and Data Analysis Lab, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
- Optical Science Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Melbourne, VIC, Australia
| | - Ronnie Blazev
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Benjamin L Parker
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Kate T Murphy
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Tarosi Senapati
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Queensland, QLD, 4072, Australia
| | - S Sean Millard
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Queensland, QLD, 4072, Australia
| | - Sarah M Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Florida, FL, 32603, USA
| | - Andrew R Judge
- Department of Physical Therapy, College of Public Health and Health Professions, University of Florida, Florida, FL, 32603, USA
| | - Lopamudra Giri
- Bioimaging and Data Analysis Lab, Department of Chemical Engineering, Indian Institute of Technology Hyderabad, Sangareddy, Telangana, India
| | - Sarah M Russell
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia
- Optical Science Centre, Faculty of Science, Engineering & Technology, Swinburne University of Technology, Hawthorn, Melbourne, VIC, Australia
- Immune Signalling Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, 3010, Australia.
| |
Collapse
|
30
|
Khan C, Rusan NM. Using Drosophila to uncover the role of organismal physiology and the tumor microenvironment in cancer. Trends Cancer 2024; 10:289-311. [PMID: 38350736 PMCID: PMC11008779 DOI: 10.1016/j.trecan.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 02/15/2024]
Abstract
Cancer metastasis causes over 90% of cancer patient fatalities. Poor prognosis is determined by tumor type, the tumor microenvironment (TME), organ-specific biology, and animal physiology. While model organisms do not fully mimic the complexity of humans, many processes can be studied efficiently owing to the ease of genetic, developmental, and cell biology studies. For decades, Drosophila has been instrumental in identifying basic mechanisms controlling tumor growth and metastasis. The ability to generate clonal populations of distinct genotypes in otherwise wild-type animals makes Drosophila a powerful system to study tumor-host interactions at the local and global scales. This review discusses advancements in tumor biology, highlighting the strength of Drosophila for modeling TMEs and systemic responses in driving tumor progression and metastasis.
Collapse
Affiliation(s)
- Chaitali Khan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Nasser M Rusan
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
31
|
Kwok SH, Liu Y, Bilder D, Kim J. Paraneoplastic renal dysfunction in fly cancer models driven by inflammatory activation of stem cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.21.586173. [PMID: 38585959 PMCID: PMC10996499 DOI: 10.1101/2024.03.21.586173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Tumors can induce systemic disturbances in distant organs, leading to physiological changes that enhance host morbidity. In Drosophila cancer models, tumors have been known for decades to cause hypervolemic 'bloating' of the abdominal cavity. Here we use allograft and transgenic tumors to show that hosts display fluid retention associated with autonomously defective secretory capacity of fly renal tubules, which function analogous to those of the human kidney. Excretion from these organs is blocked by abnormal cells that originate from inappropriate activation of normally quiescent renal stem cells (RSCs). Blockage is initiated by IL-6-like oncokines that perturb renal water-transporting cells, and trigger a damage response in RSCs that proceeds pathologically. Thus, a chronic inflammatory state produced by the tumor causes paraneoplastic fluid dysregulation by altering cellular homeostasis of host renal units.
Collapse
Affiliation(s)
- Sze Hang Kwok
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yuejiang Liu
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, 94720, USA
| | - Jung Kim
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Molecular and Cell Biology, University of California-Berkeley, Berkeley CA, 94720, USA
| |
Collapse
|
32
|
Nakamura Y, Saldajeno DP, Kawaguchi K, Kawaoka S. Progressive, multi-organ, and multi-layered nature of cancer cachexia. Cancer Sci 2024; 115:715-722. [PMID: 38254286 PMCID: PMC10921013 DOI: 10.1111/cas.16078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/27/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer cachexia is a complex, multifaceted condition that negatively impacts the health, treatment efficacy, and economic status of cancer patients. The management of cancer cachexia is an essential clinical need. Cancer cachexia is currently defined mainly according to the severity of weight loss and sarcopenia (i.e., macrosymptoms). However, such macrosymptoms may be insufficient to give clinicians clues on how to manage this condition as these symptoms appear at the late stage of cancer. We need to understand earlier events during the progression of cancer cachexia so as not to miss a clinical opportunity to control this complex syndrome. Recent research indicates that cancer-induced changes in the host are much wider than previously recognized, including disruption of liver function and the immune system. Furthermore, such changes are observed before the occurrence of visible distant metastases (i.e., in early, localized cancers). In light of these findings, we propose to expand the definition of cancer cachexia to include all cancer-induced changes to host physiology, including changes caused by early, localized cancers. This new definition of cancer cachexia can provide a new perspective on this topic, which can stimulate the research and development of novel cancer cachexia therapies.
Collapse
Affiliation(s)
- Yuki Nakamura
- Inter‐Organ Communication Research TeamInstitute for Life and Medical SciencesKyotoJapan
- Department of Breast SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Don Pietro Saldajeno
- Inter‐Organ Communication Research TeamInstitute for Life and Medical SciencesKyotoJapan
- Mathematical Informatics Laboratory, Division of Information ScienceNara Institute of Science and TechnologyIkomaNaraJapan
| | - Kosuke Kawaguchi
- Department of Breast SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | - Shinpei Kawaoka
- Inter‐Organ Communication Research TeamInstitute for Life and Medical SciencesKyotoJapan
- Department of Integrative Bioanalytics, Institute of Development, Aging and Cancer (IDAC)Tohoku UniversitySendaiJapan
| |
Collapse
|
33
|
Xu J, Liu Y, Yang F, Cao Y, Chen W, Li JSS, Zhang S, Comjean A, Hu Y, Perrimon N. Mechanistic characterization of a Drosophila model of paraneoplastic nephrotic syndrome. Nat Commun 2024; 15:1241. [PMID: 38336808 PMCID: PMC10858251 DOI: 10.1038/s41467-024-45493-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Paraneoplastic syndromes occur in cancer patients and originate from dysfunction of organs at a distance from the tumor or its metastasis. A wide range of organs can be affected in paraneoplastic syndromes; however, the pathological mechanisms by which tumors influence host organs are poorly understood. Recent studies in the fly uncovered that tumor secreted factors target host organs, leading to pathological effects. In this study, using a Drosophila gut tumor model, we characterize a mechanism of tumor-induced kidney dysfunction. Specifically, we find that Pvf1, a PDGF/VEGF signaling ligand, secreted by gut tumors activates the PvR/JNK/Jra signaling pathway in the principal cells of the kidney, leading to mis-expression of renal genes and paraneoplastic renal syndrome-like phenotypes. Our study describes an important mechanism by which gut tumors perturb the function of the kidney, which might be of clinical relevance for the treatment of paraneoplastic syndromes.
Collapse
Affiliation(s)
- Jun Xu
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China.
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Fangying Yang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Yurou Cao
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Weihang Chen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Joshua Shing Shun Li
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Shuai Zhang
- CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences, Shanghai, China
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
34
|
Yun HM, Hyun B, Song X, Hyun S. Piwi expressed in Drosophila adipose tissues regulates systemic IGF signaling and growth via IGF-binding protein. Biochem Biophys Res Commun 2024; 695:149495. [PMID: 38211532 DOI: 10.1016/j.bbrc.2024.149495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 01/07/2024] [Indexed: 01/13/2024]
Abstract
Piwi and its partner, Piwi-interacting RNA (piRNA), are pivotal in suppressing the harmful effects of transposable elements (TEs) linked to genomic insertional mutagenesis. While primarily active in Drosophila's adult gonadal tissues, causing sterility in its absence, Piwi's role in post-embryonic development remains unclear. Our study reveals Piwi's functional presence in the larval fat body, where it governs developmental growth through systemic insulin/insulin-like growth factor (IGF) signaling (IIS). Piwi knockdown in the fat body resulted in dysregulated TE expression, reduced developmental rate and body growth, and diminished systemic IIS activity. Notably, Piwi knockdown increased Imaginal Morphogenic Protein Late 2 (Imp-L2) expression, akin to insulin-like growth factor-binding protein 7 (IGFBP7), reducing systemic IIS and inhibiting body growth. This unveils a novel role for Piwi in larval adipose tissues, emphasizing its importance in regulating systemic IIS and overall organismal growth.
Collapse
Affiliation(s)
- Hyun Myoung Yun
- Department of Life Science, Chung-Ang University, Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Bom Hyun
- Department of Life Science, Chung-Ang University, Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Xinge Song
- Department of Life Science, Chung-Ang University, Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Seogang Hyun
- Department of Life Science, Chung-Ang University, Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
35
|
Wang ZY. Octopus death and dying. Integr Comp Biol 2023; 63:1209-1213. [PMID: 37437909 DOI: 10.1093/icb/icad098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 05/18/2023] [Accepted: 07/03/2023] [Indexed: 07/14/2023] Open
Affiliation(s)
- Z Yan Wang
- Department of Psychology, University of Washington, Seattle, WA 98195, USA
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
36
|
Bakopoulos D, Golenkina S, Dark C, Christie EL, Sánchez-Sánchez BJ, Stramer BM, Cheng LY. Convergent insulin and TGF-β signalling drives cancer cachexia by promoting aberrant fat body ECM accumulation in a Drosophila tumour model. EMBO Rep 2023; 24:e57695. [PMID: 38014610 DOI: 10.15252/embr.202357695] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 10/16/2023] [Accepted: 10/26/2023] [Indexed: 11/29/2023] Open
Abstract
In this study, we found that in the adipose tissue of wildtype animals, insulin and TGF-β signalling converge via a BMP antagonist short gastrulation (sog) to regulate ECM remodelling. In tumour bearing animals, Sog also modulates TGF-β signalling to regulate ECM accumulation in the fat body. TGF-β signalling causes ECM retention in the fat body and subsequently depletes muscles of fat body-derived ECM proteins. Activation of insulin signalling, inhibition of TGF-β signalling, or modulation of ECM levels via SPARC, Rab10 or Collagen IV in the fat body, is able to rescue tissue wasting in the presence of tumour. Together, our study highlights the importance of adipose ECM remodelling in the context of cancer cachexia.
Collapse
Affiliation(s)
- Daniel Bakopoulos
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Callum Dark
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | - Elizabeth L Christie
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
| | | | - Brian M Stramer
- Kings College London, Randall Centre for Cell & Molecular Biophysics, London, UK
| | - Louise Y Cheng
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Vic, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Vic, Australia
| |
Collapse
|
37
|
Krejčová G, Morgantini C, Zemanová H, Lauschke VM, Kovářová J, Kubásek J, Nedbalová P, Kamps‐Hughes N, Moos M, Aouadi M, Doležal T, Bajgar A. Macrophage-derived insulin antagonist ImpL2 induces lipoprotein mobilization upon bacterial infection. EMBO J 2023; 42:e114086. [PMID: 37807855 PMCID: PMC10690471 DOI: 10.15252/embj.2023114086] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
The immune response is an energy-demanding process that must be coordinated with systemic metabolic changes redirecting nutrients from stores to the immune system. Although this interplay is fundamental for the function of the immune system, the underlying mechanisms remain elusive. Our data show that the pro-inflammatory polarization of Drosophila macrophages is coupled to the production of the insulin antagonist ImpL2 through the activity of the transcription factor HIF1α. ImpL2 production, reflecting nutritional demands of activated macrophages, subsequently impairs insulin signaling in the fat body, thereby triggering FOXO-driven mobilization of lipoproteins. This metabolic adaptation is fundamental for the function of the immune system and an individual's resistance to infection. We demonstrated that analogically to Drosophila, mammalian immune-activated macrophages produce ImpL2 homolog IGFBP7 in a HIF1α-dependent manner and that enhanced IGFBP7 production by these cells induces mobilization of lipoproteins from hepatocytes. Hence, the production of ImpL2/IGFBP7 by macrophages represents an evolutionarily conserved mechanism by which macrophages alleviate insulin signaling in the central metabolic organ to secure nutrients necessary for their function upon bacterial infection.
Collapse
Affiliation(s)
- Gabriela Krejčová
- Department of Molecular Biology and Genetics, Faculty of ScienceUniversity of South BohemiaCeske BudejoviceCzech Republic
| | - Cecilia Morgantini
- Department of Medicine, Integrated Cardio Metabolic Center (ICMC)Karolinska InstitutetHuddingeSweden
| | - Helena Zemanová
- Department of Molecular Biology and Genetics, Faculty of ScienceUniversity of South BohemiaCeske BudejoviceCzech Republic
| | - Volker M Lauschke
- Department of Medicine, Integrated Cardio Metabolic Center (ICMC)Karolinska InstitutetHuddingeSweden
- Dr Margarete Fischer‐Bosch Institute of Clinical PharmacologyStuttgartGermany
- University of TübingenTübingenGermany
| | - Julie Kovářová
- Biology Centre CASInstitute of ParasitologyCeske BudejoviceCzech Republic
| | - Jiří Kubásek
- Department of Experimental Plant Biology, Faculty of ScienceUniversity of South BohemiaCeske BudejoviceCzech Republic
| | - Pavla Nedbalová
- Department of Molecular Biology and Genetics, Faculty of ScienceUniversity of South BohemiaCeske BudejoviceCzech Republic
| | | | - Martin Moos
- Institute of EntomologyBiology Centre CASCeske BudejoviceCzech Republic
| | - Myriam Aouadi
- Department of Medicine, Integrated Cardio Metabolic Center (ICMC)Karolinska InstitutetHuddingeSweden
| | - Tomáš Doležal
- Department of Molecular Biology and Genetics, Faculty of ScienceUniversity of South BohemiaCeske BudejoviceCzech Republic
| | - Adam Bajgar
- Department of Molecular Biology and Genetics, Faculty of ScienceUniversity of South BohemiaCeske BudejoviceCzech Republic
| |
Collapse
|
38
|
Xu W, Li G, Chen Y, Ye X, Song W. A novel antidiuretic hormone governs tumour-induced renal dysfunction. Nature 2023; 624:425-432. [PMID: 38057665 DOI: 10.1038/s41586-023-06833-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Accepted: 11/03/2023] [Indexed: 12/08/2023]
Abstract
Maintenance of renal function and fluid transport are essential for vertebrates and invertebrates to adapt to physiological and pathological challenges. Human patients with malignant tumours frequently develop detrimental renal dysfunction and oliguria, and previous studies suggest the involvement of chemotherapeutic toxicity and tumour-associated inflammation1,2. However, how tumours might directly modulate renal functions remains largely unclear. Here, using conserved tumour models in Drosophila melanogaster3, we characterized isoform F of ion transport peptide (ITPF) as a fly antidiuretic hormone that is secreted by a subset of yki3SA gut tumour cells, impairs renal function and causes severe abdomen bloating and fluid accumulation. Mechanistically, tumour-derived ITPF targets the G-protein-coupled receptor TkR99D in stellate cells of Malpighian tubules-an excretory organ that is equivalent to renal tubules4-to activate nitric oxide synthase-cGMP signalling and inhibit fluid excretion. We further uncovered antidiuretic functions of mammalian neurokinin 3 receptor (NK3R), the homologue of fly TkR99D, as pharmaceutical blockade of NK3R efficiently alleviates renal tubular dysfunction in mice bearing different malignant tumours. Together, our results demonstrate a novel antidiuretic pathway mediating tumour-renal crosstalk across species and offer therapeutic opportunities for the treatment of cancer-associated renal dysfunction.
Collapse
Affiliation(s)
- Wenhao Xu
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Gerui Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
| | - Yuan Chen
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
| | - Wei Song
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Wuhan University, Wuhan, Hubei, China.
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
39
|
Gumeni S, Lamprou M, Evangelakou Z, Manola MS, Trougakos IP. Sustained Nrf2 Overexpression-Induced Metabolic Deregulation Can Be Attenuated by Modulating Insulin/Insulin-like Growth Factor Signaling. Cells 2023; 12:2650. [PMID: 37998385 PMCID: PMC10670064 DOI: 10.3390/cells12222650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
The modulation of insulin/insulin-like growth factor signaling (IIS) is associated with altered nutritional and metabolic states. The Drosophila genome encodes eight insulin-like peptides, whose activity is regulated by a group of secreted factors, including Ecdysone-inducible gene L2 (ImpL2), which acts as a potent IIS inhibitor. We recently reported that cncC (cncC/Nrf2), the fly ortholog of Nrf2, is a positive transcriptional regulator of ImpL2, as part of a negative feedback loop aiming to suppress cncC/Nrf2 activity. This finding correlated with our observation that sustained cncC/Nrf2 overexpression/activation (cncCOE; a condition that signals organismal stress) deregulates IIS, causing hyperglycemia, the exhaustion of energy stores in flies' tissues, and accelerated aging. Here, we extend these studies in Drosophila by assaying the functional implication of ImpL2 in cncCOE-mediated metabolic deregulation. We found that ImpL2 knockdown (KD) in cncCOE flies partially reactivated IIS, attenuated hyperglycemia and restored tissue energetics. Moreover, ImpL2 KD largely suppressed cncCOE-mediated premature aging. In support, pharmacological treatment of cncCOE flies with Metformin, a first-line medication for type 2 diabetes, restored (dose-dependently) IIS functionality and extended cncCOE flies' longevity. These findings exemplify the effect of chronic stress in predisposition to diabetic phenotypes, indicating the potential prophylactic role of maintaining normal IIS functionality.
Collapse
Affiliation(s)
| | | | | | | | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Panepistimiopolis, 15784 Athens, Greece; (S.G.); (M.L.); (Z.E.); (M.S.M.)
| |
Collapse
|
40
|
Bertrand M, Szeremeta F, Hervouet-Coste N, Sarou-Kanian V, Landon C, Morisset-Lopez S, Decoville M. An adult Drosophila glioma model to highlight metabolic dysfunctions and evaluate the role of the serotonin 5-HT 7 receptor as a potential therapeutic target. FASEB J 2023; 37:e23230. [PMID: 37781977 DOI: 10.1096/fj.202300783rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/03/2023]
Abstract
Gliomas account for 50% of brain cancers and are therefore the most common brain tumors. Molecular alterations involved in adult gliomas have been identified and mainly affect tyrosine kinase receptors with amplification and/or mutation of the epidermal growth factor receptor (EGFR) and its associated signaling pathways. Several targeted therapies have been developed, but current treatments remain ineffective for glioblastomas, the most severe forms. Thus, it is a priority to identify new pharmacological targets. Drosophila glioma models established in larvae and adults are useful to identify new genes and signaling pathways involved in glioma progression. Here, we used a Drosophila glioma model in adults, to characterize metabolic disturbances associated with glioma and assess the consequences of 5-HT7 R expression on glioma development. First, by using in vivo magnetic resonance imaging, we have shown that expression of the constitutively active forms of EGFR and PI3K in adult glial cells induces brain enlargement. Then, we explored altered cellular metabolism by using high-resolution magic angle spinning NMR and 1 H-13 C heteronuclear single quantum coherence solution states. Discriminant metabolites identified highlight the rewiring of metabolic pathways in glioma and associated cachexia phenotypes. Finally, the expression of 5-HT7 R in this adult model attenuates phenotypes associated with glioma development. Collectively, this whole-animal approach in Drosophila allowed us to provide several rapid and robust phenotype readouts, such as enlarged brain volume and glioma-associated cachexia, as well as to determine the metabolic pathways involved in glioma genesis and finally to confirm the interest of the 5-HT7 R in the treatment of glioma.
Collapse
Affiliation(s)
- Marylène Bertrand
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
| | | | | | - Vincent Sarou-Kanian
- Conditions Extrêmes et Matériaux: Haute Température et Irradiation-CEMHTI-CNRS UPR 3079, Orléans, France
| | - Céline Landon
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
| | | | - Martine Decoville
- Centre de Biophysique Moléculaire-CBM, UPR 4301, CNRS, Orléans, France
- UFR Sciences et Techniques, Université d'Orléans, Orléans, France
| |
Collapse
|
41
|
Taylor J, Uhl L, Moll I, Hasan SS, Wiedmann L, Morgenstern J, Giaimo BD, Friedrich T, Alsina-Sanchis E, De Angelis Rigotti F, Mülfarth R, Kaltenbach S, Schenk D, Nickel F, Fleming T, Sprinzak D, Mogler C, Korff T, Billeter AT, Müller-Stich BP, Berriel Diaz M, Borggrefe T, Herzig S, Rohm M, Rodriguez-Vita J, Fischer A. Endothelial Notch1 signaling in white adipose tissue promotes cancer cachexia. NATURE CANCER 2023; 4:1544-1560. [PMID: 37749321 PMCID: PMC10663158 DOI: 10.1038/s43018-023-00622-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/20/2023] [Indexed: 09/27/2023]
Abstract
Cachexia is a major cause of morbidity and mortality in individuals with cancer and is characterized by weight loss due to adipose and muscle tissue wasting. Hallmarks of white adipose tissue (WAT) remodeling, which often precedes weight loss, are impaired lipid storage, inflammation and eventually fibrosis. Tissue wasting occurs in response to tumor-secreted factors. Considering that the continuous endothelium in WAT is the first line of contact with circulating factors, we postulated whether the endothelium itself may orchestrate tissue remodeling. Here, we show using human and mouse cancer models that during precachexia, tumors overactivate Notch1 signaling in distant WAT endothelium. Sustained endothelial Notch1 signaling induces a WAT wasting phenotype in male mice through excessive retinoic acid production. Pharmacological blockade of retinoic acid signaling was sufficient to inhibit WAT wasting in a mouse cancer cachexia model. This demonstrates that cancer manipulates the endothelium at distant sites to mediate WAT wasting by altering angiocrine signals.
Collapse
Affiliation(s)
- Jacqueline Taylor
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Leonie Uhl
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Theodor Boveri Institute, Department of Biochemistry and Molecular Biology, Biocenter, University of Würzburg, Würzburg, Germany
| | - Iris Moll
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sana Safatul Hasan
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Lena Wiedmann
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob Morgenstern
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
| | | | - Tobias Friedrich
- Institute of Biochemistry, University of Giessen, Giessen, Germany
- Biomedical Informatics and Systems Medicine, Science Unit for Basic and Clinical Medicine, Giessen, Germany
| | - Elisenda Alsina-Sanchis
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Francesca De Angelis Rigotti
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Ronja Mülfarth
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sarah Kaltenbach
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
| | - Darius Schenk
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Felix Nickel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine Endocrinology and Clinical Chemistry, University of Heidelberg, Heidelberg, Germany
- German Center of Diabetes Research (DZD), Neuherberg, Germany
| | - David Sprinzak
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Carolin Mogler
- Institute of Pathology, Technical University of Munich School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Korff
- Institute of Physiology and Pathophysiology, Department of Cardiovascular Physiology, University of Heidelberg, Heidelberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Adrian T Billeter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Beat P Müller-Stich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Mauricio Berriel Diaz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Tilman Borggrefe
- Institute of Biochemistry, University of Giessen, Giessen, Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
- Chair Molecular Metabolic Control, Technical University of Munich, Munich, Germany
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Center for Diabetes Research (DZD), Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Unit, Department of Inner Medicine I, Heidelberg University Hospital, Heidelberg, Germany
| | - Juan Rodriguez-Vita
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Tumor-Stroma Communication Laboratory, Centro de Investigación Príncipe Felipe, Valencia, Spain.
| | - Andreas Fischer
- Division Vascular Signaling and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Department of Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany.
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany.
| |
Collapse
|
42
|
Pranoto IKA, Lee J, Kwon YV. The roles of the native cell differentiation program aberrantly recapitulated in Drosophila intestinal tumors. Cell Rep 2023; 42:113245. [PMID: 37837622 PMCID: PMC10872463 DOI: 10.1016/j.celrep.2023.113245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 06/11/2023] [Accepted: 09/26/2023] [Indexed: 10/16/2023] Open
Abstract
Many tumors recapitulate the developmental and differentiation program of their tissue of origin, a basis for tumor cell heterogeneity. Although stem-cell-like tumor cells are well studied, the roles of tumor cells undergoing differentiation remain to be elucidated. We employ Drosophila genetics to demonstrate that the differentiation program of intestinal stem cells is crucial for enabling intestinal tumors to invade and induce non-tumor-autonomous phenotypes. The differentiation program that generates absorptive cells is aberrantly recapitulated in the intestinal tumors generated by activation of the Yap1 ortholog Yorkie. Inhibiting it allows stem-cell-like tumor cells to grow but suppresses invasiveness and reshapes various phenotypes associated with cachexia-like wasting by altering the expression of tumor-derived factors. Our study provides insight into how a native differentiation program determines a tumor's capacity to induce advanced cancer phenotypes and suggests that manipulating the differentiation programs co-opted in tumors might alleviate complications of cancer, including cachexia.
Collapse
Affiliation(s)
| | - Jiae Lee
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA
| | - Young V Kwon
- Department of Biochemistry, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
43
|
Yu K, Ramkumar N, Wong KKL, Tettweiler G, Verheyen EM. The AMPK-like protein kinases Sik2 and Sik3 interact with Hipk and induce synergistic tumorigenesis in a Drosophila cancer model. Front Cell Dev Biol 2023; 11:1214539. [PMID: 37854071 PMCID: PMC10579798 DOI: 10.3389/fcell.2023.1214539] [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: 04/30/2023] [Accepted: 09/21/2023] [Indexed: 10/20/2023] Open
Abstract
Homeodomain-interacting protein kinases (Hipks) regulate cell proliferation, apoptosis, and tissue development. Overexpression of Hipk in Drosophila causes tumorigenic phenotypes in larval imaginal discs. We find that depletion of Salt-inducible kinases Sik2 or Sik3 can suppress Hipk-induced overgrowth. Furthermore, co-expression of constitutively active forms of Sik2 or Sik3 with Hipk caused significant tissue hyperplasia and tissue distortion, indicating that both Sik2 and Sik3 can synergize with Hipk to promote tumorous phenotypes, accompanied by elevated dMyc, Armadillo/β-catenin, and the Yorkie target gene expanded. Larvae expressing these hyperplastic growths also display an extended larval phase, characteristic of other Drosophila tumour models. Examination of total protein levels from fly tissues showed that Hipk proteins were reduced when Siks were depleted through RNAi, suggesting that Siks may regulate Hipk protein stability and/or activity. Conversely, expression of constitutively active Siks with Hipk leads to increased Hipk protein levels. Furthermore, Hipk can interact with Sik2 and Sik3 by co-immunoprecipitation. Co-expression of both proteins leads to a mobility shift of Hipk protein, suggesting it is post-translationally modified. In summary, our research demonstrates a novel function of Siks in synergizing with Hipk to promote tumour growth.
Collapse
Affiliation(s)
- Kewei Yu
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Niveditha Ramkumar
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Kenneth Kin Lam Wong
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Gritta Tettweiler
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| | - Esther M. Verheyen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada
| |
Collapse
|
44
|
Saavedra P, Dumesic PA, Hu Y, Filine E, Jouandin P, Binari R, Wilensky SE, Rodiger J, Wang H, Chen W, Liu Y, Spiegelman BM, Perrimon N. REPTOR and CREBRF encode key regulators of muscle energy metabolism. Nat Commun 2023; 14:4943. [PMID: 37582831 PMCID: PMC10427696 DOI: 10.1038/s41467-023-40595-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/03/2023] [Indexed: 08/17/2023] Open
Abstract
Metabolic flexibility of muscle tissue describes the adaptive capacity to use different energy substrates according to their availability. The disruption of this ability associates with metabolic disease. Here, using a Drosophila model of systemic metabolic dysfunction triggered by yorkie-induced gut tumors, we show that the transcription factor REPTOR is an important regulator of energy metabolism in muscles. We present evidence that REPTOR is activated in muscles of adult flies with gut yorkie-tumors, where it modulates glucose metabolism. Further, in vivo studies indicate that sustained activity of REPTOR is sufficient in wildtype muscles to repress glycolysis and increase tricarboxylic acid (TCA) cycle metabolites. Consistent with the fly studies, higher levels of CREBRF, the mammalian ortholog of REPTOR, reduce glycolysis in mouse myotubes while promoting oxidative metabolism. Altogether, our results define a conserved function for REPTOR and CREBRF as key regulators of muscle energy metabolism.
Collapse
Affiliation(s)
- Pedro Saavedra
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
| | - Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Elizabeth Filine
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Patrick Jouandin
- Institut de Recherche en Cancérologie de Montpellier, INSERM, Montpellier, France
| | - Richard Binari
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
- Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Jonathan Rodiger
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Haiyun Wang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Weihang Chen
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, 02115, USA
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Howard Hughes Medical Institute, Boston, MA, 02115, USA.
| |
Collapse
|
45
|
Hsi TC, Ong KL, Sepers JJ, Kim J, Bilder D. Systemic coagulopathy promotes host lethality in a new Drosophila tumor model. Curr Biol 2023; 33:3002-3010.e6. [PMID: 37354901 PMCID: PMC11365082 DOI: 10.1016/j.cub.2023.05.071] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 04/12/2023] [Accepted: 05/31/2023] [Indexed: 06/26/2023]
Abstract
Malignant tumors trigger a complex network of inflammatory and wound repair responses, prompting Dvorak's characterization of tumors as "wounds that never heal."1 Some of these responses lead to profound defects in blood clotting, such as disseminated intravascular coagulopathy (DIC), which correlate with poor prognoses.2,3,4 Here, we demonstrate that a new tumor model in Drosophila provokes phenotypes that resemble coagulopathies observed in patients. Fly ovarian tumors overproduce multiple secreted components of the clotting cascade and trigger hypercoagulation of fly blood (hemolymph). Hypercoagulation occurs shortly after tumor induction and is transient; it is followed by a hypocoagulative state that is defective in wound healing. Cellular clotting regulators accumulate on the tumor over time and are depleted from the body, suggesting that hypocoagulation is caused by exhaustion of host clotting components. We show that rescuing coagulopathy by depleting a tumor-produced clotting factor improves survival of tumor-bearing flies, despite the fact that flies have an open (non-vascular) circulatory system. As clinical studies suggest that lethality in patients with high serum levels of clotting components can be independent of thrombotic events,5,6 our work establishes a platform for identifying alternative mechanisms by which tumor-driven coagulopathy triggers early mortality. Moreover, it opens up exploration of other conserved mechanisms of host responses to chronic wounds.
Collapse
Affiliation(s)
- Tsai-Ching Hsi
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Katy L Ong
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Jorian J Sepers
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Jung Kim
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - David Bilder
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
46
|
Li Y, Wang J, Xu Y, Meng Q, Wu M, Su Y, Miao Y, Wang Y. The water extract of Potentilla discolor Bunge (PDW) ameliorates high-sugar diet-induced type II diabetes model in Drosophila melanogaster via JAK/STAT signaling. JOURNAL OF ETHNOPHARMACOLOGY 2023:116760. [PMID: 37301307 DOI: 10.1016/j.jep.2023.116760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 06/04/2023] [Accepted: 06/07/2023] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Potentilla discolor Bunge (PD) is a member of the Rosaceae family. It has been traditionally used in folk medicine for the treatment of diabetes. Additionally, people in folk also eat fresh and tender PD stems as vegetables or brew them as tea. AIM OF THE STUDY The aim of this study was to explore the antidiabetic effects and underlying mechanisms of the water extract of Potentilla discolor (PDW) in a fruit fly model of high-sugar diet-induced type 2 diabetes. MATERIALS AND METHODS The antidiabetic efficacy of PDW was evaluated in a fruit fly model of diabetes induced by a high-sugar diet (HSD). Various physiological parameters were tested to evaluate the anti-diabetic effect of PDW. Gene expression levels related to insulin signaling pathways, glucose metabolism, lipid metabolism, and JAK/STAT signaling pathways were primarily analyzed using RT-qPCR to investigate the therapeutic mechanisms. RESULTS In this study, we found that the water extract of Potentilla discolor (PDW) can ameliorate type II diabetes phenotypes induced by the HSD in fruit flies. These phenotypes include growth rate, body size, hyperglycemia, glycogen metabolism, fat storage, and intestinal microflora homeostasis. PDW also improved the body size of s6k and rheb knockdown flies, suggesting its potential to activate the downstream insulin pathway and alleviate insulin resistance. Furthermore, we demonstrated that PDW reduced the expression of two target genes of the JAK/STAT signaling pathway, namely the insulin antagonist Impl2 and insulin receptor inhibitor Socs36E, which act as regulators inhibiting the activation of the insulin signaling pathway. CONCLUSIONS This study provides evidence for the anti-diabetic activity of PDW and suggests that its underlying mechanism may involve the improvement of insulin resistance by inhibiting the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Ying Li
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Junlin Wang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Yidong Xu
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Qinghao Meng
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Mengdi Wu
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China
| | - Yanfang Su
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China.
| | - Yaodong Miao
- Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, 300250, Tianjin, China.
| | - Yiwen Wang
- School of Pharmaceutical Science and Technology, Tianjin University, 300072, Tianjin, China.
| |
Collapse
|
47
|
Li Y, Zhou X, Cheng C, Ding G, Zhao P, Tan K, Chen L, Perrimon N, Veenstra JA, Zhang L, Song W. Gut AstA mediates sleep deprivation-induced energy wasting in Drosophila. Cell Discov 2023; 9:49. [PMID: 37221172 DOI: 10.1038/s41421-023-00541-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 03/13/2023] [Indexed: 05/25/2023] Open
Abstract
Severe sleep deprivation (SD) has been highly associated with systemic energy wasting, such as lipid loss and glycogen depletion. Despite immune dysregulation and neurotoxicity observed in SD animals, whether and how the gut-secreted hormones participate in SD-induced disruption of energy homeostasis remains largely unknown. Using Drosophila as a conserved model organism, we characterize that production of intestinal Allatostatin A (AstA), a major gut-peptide hormone, is robustly increased in adult flies bearing severe SD. Interestingly, the removal of AstA production in the gut using specific drivers significantly improves lipid loss and glycogen depletion in SD flies without affecting sleep homeostasis. We reveal the molecular mechanisms whereby gut AstA promotes the release of an adipokinetic hormone (Akh), an insulin counter-regulatory hormone functionally equivalent to mammalian glucagon, to mobilize systemic energy reserves by remotely targeting its receptor AstA-R2 in Akh-producing cells. Similar regulation of glucagon secretion and energy wasting by AstA/galanin is also observed in SD mice. Further, integrating single-cell RNA sequencing and genetic validation, we uncover that severe SD results in ROS accumulation in the gut to augment AstA production via TrpA1. Altogether, our results demonstrate the essential roles of the gut-peptide hormone AstA in mediating SD-associated energy wasting.
Collapse
Affiliation(s)
- Yingge Li
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Xiaoya Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Guangming Ding
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Peng Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Kai Tan
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Lixia Chen
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Norbert Perrimon
- Department of Genetics, Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Jan A Veenstra
- INCIA, UMR 5287 CNRS, University of Bordeaux, Talence, France
| | - Luoying Zhang
- Key Laboratory of Molecular Biophysics of Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wei Song
- Department of Hepatobiliary and Pancreatic Surgery, Medical Research Institute, Frontier Science Center of Immunology and Metabolism, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
48
|
Liu Y, Dantas E, Ferrer M, Liu Y, Comjean A, Davidson EE, Hu Y, Goncalves MD, Janowitz T, Perrimon N. Tumor Cytokine-Induced Hepatic Gluconeogenesis Contributes to Cancer Cachexia: Insights from Full Body Single Nuclei Sequencing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.15.540823. [PMID: 37292804 PMCID: PMC10245574 DOI: 10.1101/2023.05.15.540823] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
A primary cause of death in cancer patients is cachexia, a wasting syndrome attributed to tumor-induced metabolic dysregulation. Despite the major impact of cachexia on the treatment, quality of life, and survival of cancer patients, relatively little is known about the underlying pathogenic mechanisms. Hyperglycemia detected in glucose tolerance test is one of the earliest metabolic abnormalities observed in cancer patients; however, the pathogenesis by which tumors influence blood sugar levels remains poorly understood. Here, utilizing a Drosophila model, we demonstrate that the tumor secreted interleukin-like cytokine Upd3 induces fat body expression of Pepck1 and Pdk, two key regulatory enzymes of gluconeogenesis, contributing to hyperglycemia. Our data further indicate a conserved regulation of these genes by IL-6/JAK-STAT signaling in mouse models. Importantly, in both fly and mouse cancer cachexia models, elevated gluconeogenesis gene levels are associated with poor prognosis. Altogether, our study uncovers a conserved role of Upd3/IL-6/JAK-STAT signaling in inducing tumor-associated hyperglycemia, which provides insights into the pathogenesis of IL-6 signaling in cancer cachexia.
Collapse
Affiliation(s)
- Ying Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Ezequiel Dantas
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Miriam Ferrer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724 USA
| | - Yifang Liu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Aram Comjean
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Emma E. Davidson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724 USA
| | - Yanhui Hu
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Marcus D. Goncalves
- Division of Endocrinology, Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10065, USA
| | - Tobias Janowitz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY 11724 USA
- Northwell Health Cancer Institute, Northwell Health, New Hyde Park, New York, NY 11042 USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| |
Collapse
|
49
|
Okada M, Takano T, Ikegawa Y, Ciesielski H, Nishida H, Yoo SK. Oncogenic stress-induced Netrin is a humoral signaling molecule that reprograms systemic metabolism in Drosophila. EMBO J 2023:e111383. [PMID: 37140455 DOI: 10.15252/embj.2022111383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 04/01/2023] [Accepted: 04/20/2023] [Indexed: 05/05/2023] Open
Abstract
Cancer exerts pleiotropic, systemic effects on organisms, leading to health deterioration and eventually to organismal death. How cancer induces systemic effects on remote organs and the organism itself still remains elusive. Here we describe a role for NetrinB (NetB), a protein with a particularly well-characterized role as a tissue-level axon guidance cue, in mediating oncogenic stress-induced organismal, metabolic reprogramming as a systemic humoral factor. In Drosophila, Ras-induced dysplastic cells upregulate and secrete NetB. Inhibition of either NetB from the transformed tissue or its receptor in the fat body suppresses oncogenic stress-induced organismal death. NetB from the dysplastic tissue remotely suppresses carnitine biosynthesis in the fat body, which is critical for acetyl-CoA generation and systemic metabolism. Supplementation of carnitine or acetyl-CoA ameliorates organismal health under oncogenic stress. This is the first identification, to our knowledge, of a role for the Netrin molecule, which has been studied extensively for its role within tissues, in humorally mediating systemic effects of local oncogenic stress on remote organs and organismal metabolism.
Collapse
Affiliation(s)
- Morihiro Okada
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
| | - Tomomi Takano
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
| | - Yuko Ikegawa
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hanna Ciesielski
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Division of Developmental Biology and Regenerative Medicine, Kobe University, Kobe, Japan
| | - Hiroshi Nishida
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Division of Cell Physiology, Kobe University, Kobe, Japan
| | - Sa Kan Yoo
- Physiological Genetics Laboratory, RIKEN CPR, Kobe, Japan
- Laboratory for Homeodynamics, RIKEN BDR, Kobe, Japan
- Division of Developmental Biology and Regenerative Medicine, Kobe University, Kobe, Japan
| |
Collapse
|
50
|
Wang X, Li J, Zhang W, Wang F, Wu Y, Guo Y, Wang D, Yu X, Li A, Li F, Xie Y. IGFBP-3 promotes cachexia-associated lipid loss by suppressing insulin-like growth factor/insulin signaling. Chin Med J (Engl) 2023; 136:974-985. [PMID: 37014770 PMCID: PMC10278738 DOI: 10.1097/cm9.0000000000002628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Indexed: 04/05/2023] Open
Abstract
BACKGROUND Progressive lipid loss of adipose tissue is a major feature of cancer-associated cachexia. In addition to systemic immune/inflammatory effects in response to tumor progression, tumor-secreted cachectic ligands also play essential roles in tumor-induced lipid loss. However, the mechanisms of tumor-adipose tissue interaction in lipid homeostasis are not fully understood. METHODS The yki -gut tumors were induced in fruit flies. Lipid metabolic assays were performed to investigate the lipolysis level of different types of insulin-like growth factor binding protein-3 (IGFBP-3) treated cells. Immunoblotting was used to display phenotypes of tumor cells and adipocytes. Quantitative polymerase chain reaction (qPCR) analysis was carried out to examine the gene expression levels such as Acc1 , Acly , and Fasn et al . RESULTS In this study, it was revealed that tumor-derived IGFBP-3 was an important ligand directly causing lipid loss in matured adipocytes. IGFBP-3, which is highly expressed in cachectic tumor cells, antagonized insulin/IGF-like signaling (IIS) and impaired the balance between lipolysis and lipogenesis in 3T3-L1 adipocytes. Conditioned medium from cachectic tumor cells, such as Capan-1 and C26 cells, contained excessive IGFBP-3 that potently induced lipolysis in adipocytes. Notably, neutralization of IGFBP-3 by neutralizing antibody in the conditioned medium of cachectic tumor cells significantly alleviated the lipolytic effect and restored lipid storage in adipocytes. Furthermore, cachectic tumor cells were resistant to IGFBP-3 inhibition of IIS, ensuring their escape from IGFBP-3-associated growth suppression. Finally, cachectic tumor-derived ImpL2, the IGFBP-3 homolog, also impaired lipid homeostasis of host cells in an established cancer-cachexia model in Drosophila . Most importantly, IGFBP-3 was highly expressed in cancer tissues in pancreatic and colorectal cancer patients, especially higher in the sera of cachectic cancer patients than non-cachexia cancer patients. CONCLUSION Our study demonstrates that tumor-derived IGFBP-3 plays a critical role in cachexia-associated lipid loss and could be a biomarker for diagnosis of cachexia in cancer patients.
Collapse
Affiliation(s)
- Xiaohui Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Jia Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Wei Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Feng Wang
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yunzi Wu
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yulin Guo
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Dong Wang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xinfeng Yu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Ang Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yibin Xie
- Department of Pancreatic and Gastric Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|