1
|
Koyama T, Saeed U, Rewitz K, Halberg KV. The Integrative Physiology of Hormone Signaling: Insights from Insect Models. Physiology (Bethesda) 2025; 40:0. [PMID: 39887191 DOI: 10.1152/physiol.00030.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 11/18/2024] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Hormones orchestrate virtually all physiological processes in animals and enable them to adjust internal responses to meet diverse physiological demands. Studies in both vertebrates and insects have uncovered many novel hormones and dissected the physiological mechanisms they regulate, demonstrating a remarkable conservation in endocrine signaling across the tree of life. In this review, we focus on recent advances in insect research, which have provided a more integrative view of the conserved interorgan communication networks that control physiology. These new insights have been driven by experimental advantages inherent to insects, which over the past decades have aligned with new technologies and sophisticated genetic tools, to transform insect genetic models into a powerful testbed for posing new questions and exploring longstanding issues in endocrine research. Here, we illustrate how insect studies have addressed classic questions in three main areas, hormonal control of growth and development, neuroendocrine regulation of ion and water balance, and hormonal regulation of behavior and metabolism, and how these discoveries have illuminated our fundamental understanding of endocrine signaling in animals. The application of integrative physiology in insect systems to questions in endocrinology and physiology is expanding and is poised to be a crucible of discovery, revealing fundamental mechanisms of hormonal regulation that underlie animal adaptations to their environments.
Collapse
Affiliation(s)
- Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Usama Saeed
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kenneth V Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
2
|
Ukita Y, Suzuki R, Miyoshi K, Saito K, Okumura M, Chihara T. Generation of Odorant Receptor-QF2 Knock-In Drivers for Improved Analysis of Olfactory Circuits in Drosophila. Genes Cells 2025; 30:e70028. [PMID: 40439284 PMCID: PMC12121335 DOI: 10.1111/gtc.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/01/2025] [Accepted: 05/14/2025] [Indexed: 06/02/2025]
Abstract
Drosophila melanogaster has provided numerous insights into the olfactory system, primarily relying on a series of transgenic Gal4 drivers. The combined use of Gal4/UAS and a second binary expression system, such as the QF/QUAS system, provides the opportunity to manipulate the two distinct cell populations, thereby accelerating the elucidation of the olfactory neural mechanisms. However, resources apart from the Gal4/UAS system have been poorly developed. In this study, we generated a series of odorant receptor (Or)-QF2 knock-in driver (Or-QF2KI) lines for 23 Ors using the CRISPR/Cas9 knock-in method. In these lines, the QF2 protein is cotranslated with each Or product. The expression pattern of the Or-QF2KI drivers mostly corresponded to that of the Or-Gal4 drivers. In addition, the Or42a-QF2KI driver identified the additional expression pattern of Or42a, which is consistent with the data of single-nucleus RNA sequencing and is attributed to the Or-QF2KI drivers' ability to reflect the endogenous expression of the Or genes. Thus, these Or-QF2KI drivers can be used as valuable genetic tools for olfactory research in Drosophila.
Collapse
Affiliation(s)
- Yumiko Ukita
- Program of Biomedical ScienceGraduate School of Integrated Sciences for Life, Hiroshima UniversityHigashi‐HiroshimaHiroshimaJapan
| | - Ryoka Suzuki
- Program of Biomedical ScienceGraduate School of Integrated Sciences for Life, Hiroshima UniversityHigashi‐HiroshimaHiroshimaJapan
| | - Keita Miyoshi
- Department of Chromosome ScienceNational Institute of Genetics, Research Organization of Information and Systems (ROIS)ShizuokaJapan
- Graduate Institute for Advanced Studies, SOKENDAIShizuokaJapan
| | - Kuniaki Saito
- Department of Chromosome ScienceNational Institute of Genetics, Research Organization of Information and Systems (ROIS)ShizuokaJapan
- Graduate Institute for Advanced Studies, SOKENDAIShizuokaJapan
| | - Misako Okumura
- Program of Biomedical ScienceGraduate School of Integrated Sciences for Life, Hiroshima UniversityHigashi‐HiroshimaHiroshimaJapan
- Program of Basic Biology, Graduate School of Integrated Sciences for LifeHiroshima UniversityHigashi‐HiroshimaHiroshimaJapan
| | - Takahiro Chihara
- Program of Biomedical ScienceGraduate School of Integrated Sciences for Life, Hiroshima UniversityHigashi‐HiroshimaHiroshimaJapan
- Program of Basic Biology, Graduate School of Integrated Sciences for LifeHiroshima UniversityHigashi‐HiroshimaHiroshimaJapan
| |
Collapse
|
3
|
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
|
4
|
Harris DE, Kim JJ, Stern SR, Vicars HM, Matias NR, Gallicchio L, Baker CC, Fuller MT. An RNA-binding regulatory cascade controls the switch from proliferation to differentiation in the Drosophila male germ cell lineage. Proc Natl Acad Sci U S A 2025; 122:e2418279122. [PMID: 40377994 PMCID: PMC12107169 DOI: 10.1073/pnas.2418279122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 03/31/2025] [Indexed: 05/18/2025] Open
Abstract
The switch from precursor cell proliferation to onset of differentiation in adult stem cell lineages must be carefully regulated to produce sufficient progeny to maintain and repair tissues, yet prevent overproliferation that may enable oncogenesis. In the Drosophila male germ cell lineage, spermatogonia produced by germ line stem cells undergo a limited number of transit amplifying mitotic divisions before switching to the spermatocyte program that sets up meiosis and eventual spermatid differentiation. The number of transit amplifying divisions is set by accumulation of the bag-of-marbles (Bam) protein to a critical threshold. In bam mutants, spermatogonia proliferate through several extra rounds of mitosis and then die without becoming spermatocytes. Here, we show that a key role of Bam for the mitosis to differentiation switch is repressing expression of Held Out Wings (how), homolog of mammalian Quaking. Knockdown of how in germ cells was sufficient to allow spermatogonia mutant for bam or its partner benign gonial cell neoplasm to differentiate, while forced expression of nuclear-targeted How protein in spermatogonia wild-type for bam resulted in continued proliferation at the expense of differentiation. Our findings suggest that Bam targets how RNA for degradation by acting as an adapter to recruit the CCR4-NOT deadenylation complex via binding its subunit, Caf40. As How is itself an RNA-binding protein with roles in RNA processing, our findings reveal that the switch from proliferation to meiosis and differentiation in the Drosophila male germ line adult stem cell lineage is regulated by a cascade of RNA-binding proteins.
Collapse
Affiliation(s)
- Devon E. Harris
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Jongmin J. Kim
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Sarah R. Stern
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Hannah M. Vicars
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Neuza R. Matias
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Lorenzo Gallicchio
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Catherine C. Baker
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA94305
| | - Margaret T. Fuller
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA94305
- Department of Genetics, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
5
|
Wolfstetter G, Masudi T, Uçkun E, Zhu JY, Yi M, Anthonydhason V, Guan J, Sonnenberg H, Han Z, Palmer RH. Alk Tango reveals a role for Jeb/Alk signaling in the Drosophila heart. Cell Commun Signal 2025; 23:229. [PMID: 40382638 DOI: 10.1186/s12964-025-02150-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 03/11/2025] [Indexed: 05/20/2025] Open
Abstract
Anaplastic lymphoma kinase (Alk) signaling is important in a variety of biological contexts such as cell type specification, regulation of metabolic and endocrine programs, behavior, and cancer. In this work, we generated a Tango GPCR assay-based, dimerization-sensitive Alk activity reporter (AlkTango) and followed receptor activation throughout Drosophila development. AlkTango reports Alk activation in embryonic and larval tissues previously linked to Alk signaling. Remarkably, AlkTango was active in the heart of Drosophila larvae and adult flies. We show that cardiomyocytes express Alk from late embryonic stages to adulthood, while jeb expression in pericardial cells coincided with AlkTango activity. Perturbation of cardiac Alk signaling leads to decreased adult survival as well as lower fitness and increased lethality in response to heat stress. In keeping with a role for Alk, heart measurements reveal arrythmia and irregular muscle contraction upon ligand stimulation. Finally, activation of cardiac Alk signaling induces hyperplasia in the accessory wing hearts of adult flies.
Collapse
Affiliation(s)
- G Wolfstetter
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden.
| | - T Masudi
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - E Uçkun
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - J Y Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - M Yi
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - V Anthonydhason
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - J Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - H Sonnenberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Z Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - R H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden.
| |
Collapse
|
6
|
Epiney DG, Chaya GM, Dillon NR, Lai SL, Doe CQ. Single nuclei RNA-sequencing of adult brain neurons derived from type 2 neuroblasts reveals transcriptional complexity in the insect central complex. eLife 2025; 14:RP105896. [PMID: 40371710 PMCID: PMC12081001 DOI: 10.7554/elife.105896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025] Open
Abstract
In both Drosophila and mammals, the brain contains the most diverse population of cell types of any tissue. It is generally accepted that transcriptional diversity is an early step in generating neuronal and glial diversity, followed by the establishment of a unique gene expression profile that determines morphology, connectivity, and function. In Drosophila, there are two types of neural stem cells, called Type 1 (T1) and Type 2 (T2) neuroblasts. The diversity of T2-derived neurons contributes a large portion of the central complex (CX), a conserved brain region that plays a role in sensorimotor integration. Recent work has revealed much of the connectome of the CX, but how this connectome is assembled remains unclear. Mapping the transcriptional diversity of T2-derived neurons is a necessary step in linking transcriptional profile to the assembly of the adult brain. Here we perform single nuclei RNA sequencing of T2 neuroblast-derived adult neurons and glia. We identify clusters containing all known classes of glia, clusters that are male/female enriched, and 161 neuron-specific clusters. We map neurotransmitter and neuropeptide expression and identify unique transcription factor combinatorial codes for each cluster. This is a necessary step that directs functional studies to determine whether each transcription factor combinatorial code specifies a distinct neuron type within the CX. We map several columnar neuron subtypes to distinct clusters and identify two neuronal classes (NPF+ and AstA+) that both map to two closely related clusters. Our data support the hypothesis that each transcriptional cluster represents one or a few closely related neuron subtypes.
Collapse
Affiliation(s)
- Derek G Epiney
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Gonzalo Morales Chaya
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Noah R Dillon
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Sen-Lin Lai
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| | - Chris Q Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of OregonEugeneUnited States
| |
Collapse
|
7
|
Asahina K, Zelikowsky M. Comparative Perspectives on Neuropeptide Function and Social Isolation. Biol Psychiatry 2025; 97:942-952. [PMID: 39892690 PMCID: PMC12048258 DOI: 10.1016/j.biopsych.2025.01.019] [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: 08/30/2024] [Revised: 01/07/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
Chronic social isolation alters behavior across animal species. Genetic model organisms such as mice and flies provide crucial insight into the molecular and physiological effects of social isolation on brain cells and circuits. Here, we comparatively review recent findings regarding the function of conserved neuropeptides in social isolation in mice and flies. Analogous functions of 3 classes of neuropeptides-tachykinins, cholecystokinins, and neuropeptide Y/F-in the two model organisms suggest that these molecules may be involved in modulating behavioral changes induced by social isolation across a wider range of species, including humans. Comparative approaches armed with tools to dissect neuropeptidergic function can lead to an integrated understanding of the impacts of social isolation on brain circuits and behavior.
Collapse
Affiliation(s)
- Kenta Asahina
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, La Jolla, California.
| | - Moriel Zelikowsky
- Department of Neurobiology, School of Medicine, The University of Utah, Salt Lake City, Utah
| |
Collapse
|
8
|
Gillen A, Keenan S, Skov M, Akram M, Davies SA, Dow JAT. DIGITtally-a new tool for streamlining and simplifying Drosophila melanogaster meta-analysis. Nucleic Acids Res 2025:gkaf393. [PMID: 40357659 DOI: 10.1093/nar/gkaf393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Drosophila melanogaster has one of the deepest research bases within the life sciences, with a wealth of high-quality tissue- and cell type-specific transcriptomic data available. However, integrating large datasets derived from disparate sources is not trivial. We have designed a broadly applicable solution to this problem in the form of the Drosophila Interesting Genes in Individual Tissues-tally (DIGITtally) system. It is freely available online at www.digittally.org. DIGITtally is customizable and hypothesis-free, allowing meta-analysis across the Drosophila research space along with analysis of conservation in other species, querying 10 data sources for seven indicators of tissue-specific activity. We have applied DIGITtally to a pertinent question within entomology-that is, whether a specific pattern of gene expression underlies the transporting activity of epithelial tissues (an 'epitheliome'). By using DIGITtally to survey gene expression throughout the tissues comprising the D. melanogaster alimentary canal (salivary gland, midgut, Malpighian tubules, and hindgut), we have verified the existence of a specific 'epithelial' vacuolar-type ATPase configuration.
Collapse
Affiliation(s)
- Andrew Gillen
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Shannon Keenan
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Maiken Skov
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Mehwish Akram
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Shireen A Davies
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Julian A T Dow
- School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| |
Collapse
|
9
|
Huang RN, Luo SY, Huang T, Li XS, Zhou FC, Yin WH, Chen ZR, Yuan SZ, Li LY, Tang B, Qiao JD. The interaction of UBR4, LRP1, and OPHN1 in refractory epilepsy: Drosophila model to investigate the oligogenic effect on epilepsy. Neurobiol Dis 2025; 212:106955. [PMID: 40374006 DOI: 10.1016/j.nbd.2025.106955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/11/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025] Open
Abstract
Refractory epilepsy is an intractable neurological disorder that can be associated with oligogenic/polygenic etiologies. Through trio-based whole-exome sequencing analysis, we identified a clinical case of refractory epilepsy with three candidate gene variants: UBR4, LRP1, and OPHN1. Utilizing the Gal4-UAS system and double-balancer tool, we generated single, double, and triple knockdown Drosophila models to investigate the interactions of the three candidate genes. Seizure behavioral experiments combined with logistic regression analysis revealed the individual epileptogenicity and significant synergistic epileptogenic effects of the three mutations. By constructing a SHAP-XGBoost machine learning model integrating seizure behavior data with knockdown efficiency metrics, we discovered that LRP1 mutation served as the primary effector in the oligogenic system. Based on transcriptome analysis, main related processes of oxidative stress and metabolic imbalance together with expressional dysregulation separately of 48, 52, and 43 epilepsy-associated genes were discovered to confirm the epileptogenicity of OPHN1 knockdown, UBR4-LRP1 knockdown, and UBR4-LRP1-OPHN1 knockdown. Up-regulation of COX7AL and ND-B8 enriched in metabolic pathways and down-regulation of Diedel enriched in extracellular space component were indicated to be responsible for the significant epileptogenicity of the oligogenic knockdown. For this clinical instance, epileptic pharmacoresistance was considered to be triggered by a combination of KIF gene family, SLC gene family, and ASIC gene family. This study established a novel framework to clarify the multiple genetic structure of epileptogenicity in refractory epilepsy with oligogenic background, which could be critical to translational medicine and precision therapy development.
Collapse
Affiliation(s)
- Rui-Na Huang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Changgang Dong Road, Guangzhou 510000, China; The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou 510000, China
| | - Si-Yuan Luo
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou 510000, China
| | - Tao Huang
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou 510000, China
| | - Xiong-Sheng Li
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou 510000, China
| | - Fan-Chao Zhou
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou 510000, China
| | - Wei-Hao Yin
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou 510000, China
| | - Ze-Ru Chen
- The Second Clinical Medicine School, Guangzhou Medical University, Guangzhou 510000, China
| | - Shi-Zhan Yuan
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Changgang Dong Road, Guangzhou 510000, China
| | - Ling-Ying Li
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Changgang Dong Road, Guangzhou 510000, China
| | - Bin Tang
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Changgang Dong Road, Guangzhou 510000, China.
| | - Jing-Da Qiao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital of Guangzhou Medical University, Changgang Dong Road, Guangzhou 510000, China.
| |
Collapse
|
10
|
Park YJ, Lu TC, Jackson T, Goodman LD, Ran L, Chen J, Liang CY, Harrison E, Ko C, Chen X, Wang B, Hsu AL, Ochoa E, Bieniek KF, Yamamoto S, Zhu Y, Zheng H, Qi Y, Bellen HJ, Li H. Distinct systemic impacts of Aβ42 and Tau revealed by whole-organism snRNA-seq. Neuron 2025:S0896-6273(25)00299-5. [PMID: 40381615 DOI: 10.1016/j.neuron.2025.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/27/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025]
Abstract
Both neuronal and peripheral tissues become disrupted in Alzheimer's disease (AD). However, a comprehensive understanding of how AD impacts different tissues across the whole organism is lacking. Using Drosophila, we generated an AD Fly Cell Atlas (AD-FCA) based on whole-organism single-nucleus transcriptomes of 219 cell types from flies expressing AD-associated proteins, either human amyloid-β 42 peptide (Aβ42) or Tau, in neurons. We found that Aβ42 primarily affects the nervous system, including sensory neurons, while Tau induces accelerated aging in peripheral tissues. We identified a neuronal cluster enriched in Aβ42 flies, which has high lactate dehydrogenase (LDH) expression. This LDH-high cluster is conserved in 5XFAD mouse and human AD datasets. We found a conserved defect in fat metabolism from both fly and mouse tauopathy models. The AD-FCA offers new insights into how Aβ42 or Tau systemically and differentially affects a whole organism and provides a valuable resource for understanding brain-body communication in neurodegeneration.
Collapse
Affiliation(s)
- Ye-Jin Park
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tzu-Chiao Lu
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Tyler Jackson
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Program in Cancer Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA
| | - Lindsey Ran
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jiaye Chen
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chung-Yi Liang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Erin Harrison
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christina Ko
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Xi Chen
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Baiping Wang
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ao-Lin Hsu
- Institute of Biochemistry and Molecular Biology, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan; Department of Internal Medicine, Division of Geriatric and Palliative Medicine, University of Michigan, Ann Arbor, MI 28109, USA
| | - Elizabeth Ochoa
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Kevin F Bieniek
- Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, UT Health San Antonio, San Antonio, TX 78229, USA; Department of Pathology & Laboratory Medicine, UT Health San Antonio, San Antonio, TX 78229, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yi Zhu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hui Zheng
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yanyan Qi
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA; Program in Development, Disease Models & Therapeutics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Hongjie Li
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
11
|
Yu J, Huang Q, Fu Y, He L, Shen C, Chen X, Li Z, Li J, Wang C, Wang X, Yang B, Lin Z, Qiao C, Tan X, Yang X, Chen H, Zheng Y, Zheng B, Sun F. Multi-omics integration reveals Vha68-3 as a testicular aging-specific factor that coordinates spermatid elongation through mitochondrial metabolic homeostasis. Cell Mol Biol Lett 2025; 30:58. [PMID: 40346547 PMCID: PMC12065321 DOI: 10.1186/s11658-025-00737-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Testicular aging has profound effects on spermatogenesis, sperm function, and the spermatogenic microenvironment, contributing to reduced male fertility. However, the precise molecular mechanisms by which mitochondria influence spermiogenesis during aging still remain largely unclear. METHODS Vha68-3 KO flies were generated using the CRISPR/Cas9 technique. Testicular phenotypes and functions were mainly observed through immunofluorescence staining and transmission electron microscopy. Multi-omics study was mainly conducted through single-cell RNA sequencing and transcriptome-metabolomics association analysis. Vha68-3 binding proteins were identified via liquid chromatography-tandem mass spectrometry. The therapeutic potential of modulating mitochondrial metabolism for testicular aging mainly relied on the dietary intake of related compounds in fruit flies. RESULTS In this study, we identified Vha68-3, a testis-specific subunit of the V-type adenosine triphosphate (ATP) synthase, predominantly localized in the tails of elongated spermatids, as a key age-related regulator of male fertility and spermatid elongation in Drosophila testes. Crucially, Vha68-3 deficiency impaired mitochondrial homeostasis in elongated spermatids during testicular aging. Through a multi-omics approach, including single-cell transcriptomics, protein interaction mapping of Vha68-3, and transcriptome-metabolome integration, we identified pyruvate metabolism as a critical pathway disrupted by Vha68-3 deficiency. Moreover, dietary supplementation with pyruvate (PA), S-lactoylglutathione (SLG), and phosphoenolpyruvate (PEP) effectively alleviated mitochondrial dysfunction and testicular aging linked to Vha68-3 deficiency. CONCLUSIONS Our findings uncover novel mechanisms by which mitochondrial metabolism regulates spermatid elongation and propose potential therapeutic strategies to combat mitochondrial metabolic disorders in aging testes.
Collapse
Affiliation(s)
- Jun Yu
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China.
| | - Qiuru Huang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Yangbo Fu
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Lei He
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Cong Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, China
| | - Xia Chen
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Zhiran Li
- Department of Andrology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Jiaxin Li
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Chenyu Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Xinda Wang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Binbin Yang
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Ziwen Lin
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China
| | - Chen Qiao
- Department of Clinical Pharmacy, The Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, 212001, China
| | - Xiaofang Tan
- Reproductive Medicine Center, The Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, 226001, China
| | - Xiaoqing Yang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, China
| | - Hao Chen
- Guangzhou Women and Children's Medical Center, GMU-GIBH Joint School of Life Science, The Guangdong-Hong Kong-Macao Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, 511495, China
| | - Ying Zheng
- Department of Histology and Embryology, School of Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Bo Zheng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproduction and Genetics, Suzhou Municipal Hospital, Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, 215002, China.
| | - Fei Sun
- Institute of Reproductive Medicine, Medical School, Nantong University, Nantong, 226001, China.
- School of Basic Medical Sciences, Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, 750004, China.
| |
Collapse
|
12
|
Cridland JM, Polston ES, Begun DJ. New perspectives on Drosophila melanogaster de novo gene origination revealed by investigation of ancient African genetic variation. Genetics 2025; 230:iyaf044. [PMID: 40106667 PMCID: PMC12059636 DOI: 10.1093/genetics/iyaf044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/04/2025] [Indexed: 03/22/2025] Open
Abstract
De novo genes can be defined as sequences producing evolutionarily derived transcripts that are not homologous to transcripts produced in an ancestor. While they appear to be taxonomically widespread, there is little agreement regarding their abundance, their persistence times in genomes, the population genetic processes responsible for their spread or loss, or their possible functions. In Drosophila melanogaster, 2 approaches have been used to discover these genes and investigate their properties. One uses traditional comparative approaches and existing genomic resources and annotations. A second approach uses raw transcriptome data to discover unannotated genes for which there is no evidence of presence in related species. Investigations using the second approach have focused on D. melanogaster genotypes from recently established cosmopolitan populations. However, most of the genetic variation in the species is found in African populations, suggesting the possibility that fuller understanding of genetic novelties in the species may follow from studies of these populations. Here, we investigate de novo gene candidates expressed in testis and accessory glands in a sample of flies from Zambia and compare them with candidate de novo genes expressed in North American populations. We report a large number of previously undiscovered de novo gene candidates, most of which are expressed polymorphically. Many are predicted to code for secreted proteins. In spite of much different levels of genomic variation in Zambian and North American populations, they express similar numbers of candidate de novo genes. We find evidence from genetic analysis of Raleigh inbred lines that a fraction of rarely expressed gene candidates in this population represent deleterious transcription promoted by inbreeding depression. Many de novo gene candidates are expressed in multiple tissues and both sexes, raising questions about how they may interact with natural selection. The relative importance of positive and negative selection, however, remains unclear.
Collapse
Affiliation(s)
- Julie M Cridland
- Department of Evolution and Ecology, University of California, Davis, Davis, CA 95616, USA
| | - Elizabeth S Polston
- Department of Evolution and Ecology, University of California, Davis, Davis, CA 95616, USA
| | - David J Begun
- Department of Evolution and Ecology, University of California, Davis, Davis, CA 95616, USA
| |
Collapse
|
13
|
Messer CL, Burghardt E, McDonald JA. A deficiency screen of the X chromosome for Rap1 GTPase dominant interacting genes in Drosophila border cell migration. G3 (BETHESDA, MD.) 2025; 15:jkaf040. [PMID: 39993182 PMCID: PMC12060239 DOI: 10.1093/g3journal/jkaf040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025]
Abstract
Collective cell migration is critical to embryonic development, wound healing, and the immune response, but also drives tumor dissemination. Understanding how cell collectives coordinate migration in vivo has been a challenge, with potential therapeutic benefits that range from addressing developmental defects to designing targeted cancer treatments. The small GTPase Rap1 has emerged as a regulator of both embryogenesis and cancer cell migration. How active Rap1 coordinates downstream signaling functions required for coordinated collective migration is poorly understood. Drosophila border cells undergo a stereotyped and genetically tractable in vivo migration within the developing egg chamber of the ovary. This group of 6-8 cells migrates through a densely packed tissue microenvironment and serves as an excellent model for collective cell migration during development and disease. Rap1, like all small GTPases, has distinct activity state switches that link extracellular signals to organized cell behaviors. Proper regulation of Rap1 activity is essential for successful border cell migration yet the signaling partners and other downstream effectors are poorly characterized. Using the known requirement for Rap1 in border cell migration, we conducted a dominant suppressor screen for genes whose heterozygous loss modifies the migration defects observed upon constitutively active Rap1V12 expression. Here, we identified 7 genomic regions on the X chromosome that interact with Rap1V12. We mapped three genomic regions to single Rap1-interacting genes, frizzled 4, Ubiquitin-specific protease 16/45, and strawberry notch. Thus, this unbiased screening approach identified multiple new candidate regulators of Rap1 activity with roles in collective border cell migration.
Collapse
Affiliation(s)
- C Luke Messer
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
- Department of Natural Sciences, The University of Virginia's College at Wise, Wise, VA 24293, USA
| | - Emily Burghardt
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | | |
Collapse
|
14
|
Zike AB, Abel MG, Fleck SA, DeWitt ED, Weaver LN. Estrogen-related receptor is required in adult Drosophila females for germline stem cell maintenance. Dev Biol 2025; 524:132-143. [PMID: 40348318 DOI: 10.1016/j.ydbio.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
Adult tissue function is dependent on intrinsic factors that mediate stem cell self-renewal and proliferation in response to changes in physiology and the environment. The estrogen-related receptor (ERR) subfamily of orphan nuclear receptors are major transcriptional regulators of metabolism and animal physiology. In mammals, ERRs (NR3B1, NR3B2, NR3B3) have roles in regulating mitochondrial biosynthesis, lipid metabolism, as well as stem cell maintenance. The sole Drosophila ERR ortholog promotes larval growth by establishing a metabolic state during the latter half of embryogenesis. In addition, ERR is required in adult Drosophila males to coordinate glycolytic metabolism with lipid synthesis and within the testis to regulate spermatogenesis gene expression and fertility. Despite extensive work characterizing the role of ERR in Drosophila metabolism, whether ERR has a conserved requirement in regulating stem cell behavior has been understudied. To determine whether ERR regulates stem cell activity in Drosophila, we used the established adult female germline stem cell (GSC) lineage as a model. We found that whole-body ERR knockout in adult females using conditional heat shock-driven FLP-FRT recombination significantly decreases GSC number and glycolytic enzyme expression in GSCs. In addition, we found that ERR activity is required cell-autonomously in the adult female germline for maintenance of GSCs; whereas ERR regulation of GSCs is independent of its activity in adult female adipocytes. Our results highlight an ancient and conserved role for ERRs in the regulation of stem cell self-renewal.
Collapse
Affiliation(s)
- Anna B Zike
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Madison G Abel
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Sophie A Fleck
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Emily D DeWitt
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA
| | - Lesley N Weaver
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA.
| |
Collapse
|
15
|
Lau F, Binacchi R, Brugnara S, Cumplido-Mayoral A, Savino SD, Khan I, Orso A, Sartori S, Bellosta P, Carl M, Poggi L, Provenzano G. Using Single-Cell RNA sequencing with Drosophila, Zebrafish, and mouse models for studying Alzheimer's and Parkinson's disease. Neuroscience 2025; 573:505-517. [PMID: 40154937 DOI: 10.1016/j.neuroscience.2025.03.042] [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: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Alzheimer's and Parkinson's disease are the most common neurodegenerative diseases, significantly affecting the elderly with no current cure available. With the rapidly aging global population, advancing research on these diseases becomes increasingly critical. Both disorders are often studied using model organisms, which enable researchers to investigate disease phenotypes and their underlying molecular mechanisms. In this review, we critically discuss the strengths and limitations of using Drosophila, zebrafish, and mice as models for Alzheimer's and Parkinson's research. A focus is the application of single-cell RNA sequencing, which has revolutionized the field by providing novel insights into the cellular and transcriptomic landscapes characterizing these diseases. We assess how combining animal disease modeling with high-throughput sequencing and computational approaches has advanced the field of Alzheimer's and Parkinson's disease research. Thereby, we highlight the importance of integrative multidisciplinary approaches to further our understanding of disease mechanisms and thus accelerating the development of successful therapeutic interventions.
Collapse
Affiliation(s)
- Frederik Lau
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Rebecca Binacchi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Samuele Brugnara
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Alba Cumplido-Mayoral
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Serena Di Savino
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Ihsanullah Khan
- Department of Civil, Environmental and Mechanical Engineering, University of Trento 38123 Trento, Italy
| | - Angela Orso
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Samuele Sartori
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy; Department of Medicine NYU Grossman School of Medicine, 550 First Avenue, 10016 NY, USA.
| | - Matthias Carl
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy.
| | - Lucia Poggi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy.
| | - Giovanni Provenzano
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento 38123 Trento, Italy.
| |
Collapse
|
16
|
Zheng Y, Lee YC, Wang YT, Chiang PK, Chang SL, Hsu HJ, Hsu LS, Bach EA, Tseng CY. Age-related declines in niche self-renewal factors controls testis aging and spermatogonial stem cell competition through Hairless, Imp, and Chinmo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.05.01.651651. [PMID: 40370955 PMCID: PMC12077873 DOI: 10.1101/2025.05.01.651651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Aging is associated with progressive tissue decline and shifts in stem cell clonality. The role of niche signals in driving these processes remains poorly understood. Using the Drosophila testis, we identify a regulatory axis in which age-related decline of niche signals (BMPs) lead to upregulation of the co-repressor Hairless, which downregulates the RNA-binding protein Imp in aged germline stem cells (GSCs). Reduced Imp causes loss of Chinmo, a key factor in GSC aging and competition. Reduced Chinmo causes ectopic Perlecan secretion which accumulates in the testis lumen and causes GSC loss. Aging of the testis is reversed by increasing BMPs in the niche, or by overexpressing Imp or depleting Hairless in GSCs. Furthermore, GSC clones with reduced Imp or increased Hairless are more competitive, expelling wild-type neighbors and monopolizing the niche. Thus, BMPs regulate testicular niche aging through the Hairless-Imp-Chinmo axis and "winning" GSCs usurp these aging mechanisms. Highlights Aged niche cells produce less BMPs, resulting in more Hairless (H) in aged GSCs Elevated H represses Imp , resulting in less Chinmo and in ectopic ECM secretion Aging is prevented by higher BMP in niche cells, or by higher Imp or lower H in GSCs GSCs with low Imp or high H exploit these aging mechanisms to colonize the GSC pool.
Collapse
|
17
|
Lee SYJ, Dallmann CJ, Cook A, Tuthill JC, Agrawal S. Divergent neural circuits for proprioceptive and exteroceptive sensing of the Drosophila leg. Nat Commun 2025; 16:4105. [PMID: 40316553 PMCID: PMC12048489 DOI: 10.1038/s41467-025-59302-3] [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: 11/23/2024] [Accepted: 04/15/2025] [Indexed: 05/04/2025] Open
Abstract
Somatosensory neurons provide the nervous system with information about mechanical forces originating inside and outside the body. Here, we use connectomics from electron microscopy to reconstruct and analyze neural circuits downstream of the largest somatosensory organ in the Drosophila leg, the femoral chordotonal organ (FeCO). The FeCO has been proposed to support both proprioceptive sensing of the fly's femur-tibia joint and exteroceptive sensing of substrate vibrations, but it was unknown which sensory neurons and central circuits contribute to each of these functions. We found that different subtypes of FeCO sensory neurons feed into distinct proprioceptive and exteroceptive pathways. Position- and movement-encoding FeCO neurons connect to local leg motor control circuits in the ventral nerve cord (VNC), indicating a proprioceptive function. In contrast, signals from the vibration-encoding FeCO neurons are integrated across legs and transmitted to mechanosensory regions in the brain, indicating an exteroceptive function. Overall, our analyses reveal the structure of specialized circuits for processing proprioceptive and exteroceptive signals from the fly leg. These findings are consistent with a growing body of work in invertebrate and vertebrate species demonstrating the existence of specialized limb mechanosensory pathways for sensing external vibrations.
Collapse
Affiliation(s)
- Su-Yee J Lee
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Chris J Dallmann
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
- Neurobiology and Genetics, Julius-Maximilians-University of Würzburg, Würzburg, Germany
| | - Andrew Cook
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA
| | - John C Tuthill
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA.
| | - Sweta Agrawal
- Department of Neurobiology and Biophysics, University of Washington, Seattle, WA, USA.
- School of Neuroscience, Virginia Tech, Blacksburg, VA, USA.
| |
Collapse
|
18
|
Biswas P, Bako JA, Liston JB, Yu H, Wat LW, Miller CJ, Gordon MD, Huan T, Stanley M, Rideout EJ. Insulin/insulin-like growth factor signaling pathway promotes higher fat storage in Drosophila females. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.11.18.623936. [PMID: 40342968 PMCID: PMC12060994 DOI: 10.1101/2024.11.18.623936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
In Drosophila , adult females store more fat than males. While the mechanisms that restrict body fat in males are becoming clearer, less is known about how females achieve higher fat storage. Here, we perform a detailed investigation of the mechanisms that promote higher fat storage in females. We show greater intake of dietary sugar supports higher fat storage due to female-biased remodeling of the fat body lipidome. Dietary sugar stimulates a female-specific increase in Drosophila insulin-like peptide 3 (Dilp3), which acts together with greater peripheral insulin sensitivity to augment insulin/insulin-like growth factor signaling pathway (IIS) activity in adult females. Indeed, Dilp3 overexpression prevented the female-biased decrease in body fat after removal of dietary sugar. Given that adult-specific IIS inhibition caused a female-biased decrease in body fat, our data reveal IIS as a key determinant of female fat storage.
Collapse
|
19
|
Diaz AV, Tekin I, Reis T. Drosophila as a Genetic Model System to Study Organismal Energy Metabolism. Biomolecules 2025; 15:652. [PMID: 40427545 PMCID: PMC12108566 DOI: 10.3390/biom15050652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/21/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
Metabolism is the essential process by which an organism converts nutrients into energy to fuel growth, development, and repair. Metabolism at the level of a multicellular, multi-organ animal is inherently more complex than metabolism at the single-cell level. Indeed, each organ also must maintain its own homeostasis to function. At all three scales, homeostasis is a defining feature: as energy sources and energetic demands wax and wane, the system must be robust. While disruption of organismal energy homeostasis can be manifested in different ways in humans, obesity (defined as excess body fat) is an increasingly common outcome of metabolic imbalance. Here we will discuss the genetic basis of metabolic dysfunction that underlies obesity. We focus on what we are learning from Drosophila melanogaster as a model organism to explore and dissect genetic causes of metabolic dysfunction in the context of a whole organism.
Collapse
Affiliation(s)
| | | | - Tânia Reis
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
20
|
Guan C, Otchere A, Laskovs M, Papatheodorou I, Slack C. Genetic and Pharmacological Inhibition of Metabotropic Glutamate Receptor Signalling Extends Lifespan in Drosophila. Aging Cell 2025; 24:e14500. [PMID: 39943697 PMCID: PMC12073928 DOI: 10.1111/acel.14500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 05/15/2025] Open
Abstract
Invertebrate models have been instrumental in advancing our understanding of the molecular mechanisms of ageing. The isolation of single gene mutations that both extend lifespan and improve age-related health have identified potential targets for therapeutic intervention to alleviate age-related morbidity. Here, we find that genetic loss of function of the G protein-coupled metabotropic glutamate receptor (DmGluRA) in Drosophila extends the lifespan of female flies. This longevity phenotype was accompanied by lower basal levels of oxidative stress and improved stress tolerance, and differences in early-life behavioural markers. Gene expression changes in DmGluRA mutants identified reduced ribosome biogenesis, a hallmark of longevity, as a key process altered in these animals. We further show that the pro-longevity effects of reduced DmGluRA signalling are dependent on the fly homologue of Fragile X Mental Retardation Protein (FMRP), an important regulator of ribosomal protein translation. Importantly, we can recapitulate lifespan extension using a specific pharmacological inhibitor of mGluR activity. Hence, our study identifies metabotropic glutamate receptors as potential targets for age-related therapeutics.
Collapse
Affiliation(s)
- Cui Guan
- College of Health and Life SciencesAston UniversityBirminghamUK
- School of Life SciencesWarwick UniversityCoventryUK
| | - Abigail Otchere
- College of Health and Life SciencesAston UniversityBirminghamUK
| | - Mihails Laskovs
- College of Health and Life SciencesAston UniversityBirminghamUK
- School of Life SciencesWarwick UniversityCoventryUK
| | | | - Cathy Slack
- College of Health and Life SciencesAston UniversityBirminghamUK
- School of Life SciencesWarwick UniversityCoventryUK
| |
Collapse
|
21
|
Geier B, Roy B, Reiter LT. Small molecule ion channel agonist/antagonist screen reveals seizure suppression via glial Irk2 activation in a Drosophila model of Dup15q syndrome. Neurobiol Dis 2025; 208:106882. [PMID: 40122181 PMCID: PMC12117380 DOI: 10.1016/j.nbd.2025.106882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025] Open
Abstract
The neurogenetic disorder duplication 15q syndrome (Dup15q) is characterized by a high incidence of autism spectrum disorder (ASD) and pharmacoresistant epilepsy. Standard-of-care broad-spectrum anti-seizure medications (ASM) often fail to control seizures in Dup15q, emphasizing the need for the identification of new therapeutic compounds. Previously, we generated a model of Dup15q in Drosophila melanogaster by overexpressing Dube3a in glial cells, instead of neurons. This model recapitulates the spontaneous seizures present in Dup15q patients. Here, we screened a set of FDA-approved compounds for their ability to suppress seizures in repo > Dube3a flies. We used 72 compounds from the Enzo SCREEN-WELL Ion Channel Library for primary screening of seizure suppression. Six compounds were identified that significantly reduced seizure duration. Furthermore, the compounds that passed the primary and secondary screenings were associated with K+ channels. Glial-specific knockdown of the inward rectifying potassium (Irk) 2 channel exacerbated the seizure phenotype in these animals indicating a mechanism of action for drugs that bind irk2, like minoxidil, and can suppress seizures through the rebalancing of K+ extracellularly. This pharmacological and molecular investigation further supports the role of extracellular K+ content in Dup15q seizure activation and provides a putative target for therapeutic intervention.
Collapse
Affiliation(s)
- Benjamin Geier
- Department of Physiology, Tulane University, New Orleans, LA, USA; Graduate Program in Neuroscience, Tulane University, New Orleans, LA, USA
| | - Bidisha Roy
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA
| | | |
Collapse
|
22
|
Roy PR, Link N. Loss of Neuronal Imp Contributes to Seizure Behavior through Syndecan Function. eNeuro 2025; 12:ENEURO.0545-24.2025. [PMID: 40258646 PMCID: PMC12052222 DOI: 10.1523/eneuro.0545-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/23/2025] Open
Abstract
Seizures affect a large proportion of the global population and occur due to abnormal neuronal activity in the brain. Unfortunately, widespread genetic and phenotypic heterogeneity contributes to insufficient treatment options. It is critical to identify the genetic underpinnings of how seizures occur to better understand seizure disorders and improve therapeutic development. We used the Drosophila melanogaster model to identify that IGF-II mRNA-binding protein (Imp) is linked to the onset of this phenotype. Specific reduction of Imp in neurons causes seizures after mechanical stimulation. Importantly, gross motor behavior is unaffected, showing Imp loss does not affect general neuronal activity. Developmental loss of Imp is sufficient to cause seizures in adults; thus, Imp-modulated neuron development affects mature neuronal function. Since Imp is an RNA-binding protein, we sought to identify the mRNA target that Imp regulates in neurons to ensure proper neuronal activity after mechanical stress. We find that the Imp protein binds Syndecan (Sdc) mRNA, and the reduction of Sdc also causes mechanically induced seizures. Expression of Sdc in Imp-deficient neurons rescues seizure defects, showing that Sdc is sufficient to restore normal behavior after mechanical stress. We suggest that the Imp protein binds Sdc mRNA in neurons, and this functional interaction is important for normal neuronal biology and animal behavior in a mechanically induced seizure model. Since Imp and Sdc are conserved, our work highlights a neuronal-specific pathway that might contribute to seizure disorder when mutated in humans.
Collapse
Affiliation(s)
- Paula R Roy
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| | - Nichole Link
- Department of Neurobiology, University of Utah, Salt Lake City, Utah 84112
| |
Collapse
|
23
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
24
|
Lin SY, Levine MT. Paternal effects on telomere integrity during the sperm-to-embryo transition. Curr Opin Genet Dev 2025; 93:102348. [PMID: 40286639 DOI: 10.1016/j.gde.2025.102348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 03/25/2025] [Accepted: 03/30/2025] [Indexed: 04/29/2025]
Abstract
Telomeres are essential nucleoprotein structures that preserve our terminal DNA sequence and protect chromosome ends from fusion. Our vast knowledge of telomeres comes almost entirely from studies of healthy and diseased somatic cells. However, building evidence suggests that the molecules and mechanisms required for telomere integrity in somatic cells are insufficient to preserve telomere integrity during the sperm-to-embryo transition. Here, we review this growing body of work on telomere 'paternal effects', wherein zygotic telomere integrity is determined not by the genotype of the zygote but instead by the genotype of the father. Direct inheritance of sperm-specific proteins establishes paternal telomere epigenetic identity, while direct inheritance of sperm telomere length contributes to telomere length inheritance. Together, these investigations of telomere integrity through the sperm-to-embryo transition reveal potent paternal effects on zygotic telomere functions, with implications for human infertility.
Collapse
Affiliation(s)
- Sung-Ya Lin
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Mia T Levine
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
25
|
Barraza D, Ding X, Wang Z, Jugder BE, Watnick PI. The Drosophila G protein-coupled receptor, GulpR, is essential for lipid mobilization in response to nutrient-limitation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.19.649675. [PMID: 40313923 PMCID: PMC12045340 DOI: 10.1101/2025.04.19.649675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Enteroendocrine cells (EECs) of the intestinal epithelium are major regulators of metabolism and energy homeostasis. This is mainly due to their expression and secretion of enteroendocrine peptides (EEPs). These peptides serve as hormones that control many aspects of metabolic homeostasis including feeding behavior, intestinal contractions, and utilization of energy stores. Regulation of EEP production and release depends largely on EEC-exclusive G protein-coupled receptors (GPCRs) that sense nutrient levels. Here we report the identification of a GPCR expressed principally in EECs, which we have named GulpR due to its role in the response to nutrient stress. We show that GulpR regulates transcription of the EEP Tachykinin (Tk) and that both GulpR and Tk are essential for the transcriptional response that promotes survival of nutrient limitation. Infection with V. cholerae also activates transcription of Tk and lipid mobilization genes. While GulpR is required for activation of Tk transcription during infection, Tk does not play a role in regulation of lipid mobilization genes or survival of infection. Our findings identify a role for GulpR and Tk in survival of starvation and suggest that, although starvation and infection both require significant mobilization of energy stores, the signal transduction systems that regulate the metabolic response to each are distinct. Author Summary Humans and other animals, including Drosophila , metabolize dietary nutrients such as sugars, lipids, and proteins into polysaccharides, fatty acids, and amino acids, respectively, to generate energy that fuels essential cellular processes like cell division, ion transport, muscle contraction, and more. The ability to adapt to changes in nutrient availability and energy demand is therefore crucial for homeostasis and survival. Nutrient scarcity during starvation and an increased demand for energy during an immune response against pathogenic infection require utilization of the body's own lipid and glycogen stores. This adaptive response largely relies on the ability of the intestine to sense and respond to a variety of stimuli, including microbes and dietary nutrients. Here, we have identified and characterized a Drosophila melanogaster receptor that is expressed in a rare intestinal cell type. We report that this receptor regulates production of peptide hormones that are known to impact metabolic homeostasis and discover that one of these peptide hormones is crucial for utilization of systemic lipid stores when flies experience starvation but not infection stress. Our findings therefore indicate that activation of lipid mobilization in response to nutrient limitation and infection are regulated via different mechanisms.
Collapse
|
26
|
Huang Y, Xiang Z, Xiang Y, Pan H, He M, Guo Z, Kanca O, Liu C, Zhang Z, Zhan H, Wang Y, Bai QR, Bellen HJ, Wang H, Bian S, Mao X. Biallelic MED16 variants disrupt neural development and lead to an intellectual disability syndrome. J Genet Genomics 2025:S1673-8527(25)00113-4. [PMID: 40254158 DOI: 10.1016/j.jgg.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
Mediator Complex Subunit 16 (MED16, MIM: 604062) is a member of the Mediator complex which controls many aspects of transcriptional activity in all eukaryotes. Here, we report two individuals from a non-consanguineous family with biallelic variants in MED16 identified by exome sequencing. The affected individuals present with global developmental delay, intellectual disability, and dysmorphisms. To assess the pathogenicity of the variants, functional studies were performed in Drosophila and patient-derived cells. The fly ortholog med16 is expressed in neurons and some glia of the developing central nervous system (CNS). Loss of med16 leads to a reduction in eclosion and lifespan, as well as impaired synaptic transmission. In neurons differentiated from the patient-derived induced pluripotent stem cells (iPSCs), the neurite outgrowth is impaired and rescued by expression of exogenous MED16. The patient-associated variants behave as loss-of-function (LoF) alleles in flies and iPSCs. Additionally, the transcription of genes related to neuronal maturation and function is preferentially altered in patient cells relative to differentiated H9 controls. In summary, our findings support that MED16 is important for appropriate development and function, and that biallelic MED16 variants cause a neurodevelopmental disease.
Collapse
Affiliation(s)
- Yan Huang
- Department of Medical Genetics & Pediatric Research Institute, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, (Hunan Children's Hospital), Changsha, Hunan 410007, China
| | - Zhenglong Xiang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Yaqin Xiang
- Department of Medical Genetics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, (Hunan Children's Hospital), Changsha, Hunan 410007, China
| | - Hu Pan
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, Hunan 410008, China
| | - Mei He
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, Hunan 410008, China
| | - Zhenming Guo
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Chen Liu
- Neonatology, Children's Hospital Affiliated to Shandong University, Jinan, Shandong 250022, China
| | - Zhao Zhang
- Department of Medical Genetics & Pediatric Research Institute, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, (Hunan Children's Hospital), Changsha, Hunan 410007, China
| | - Huaizhe Zhan
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Yuan Wang
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Qing-Ran Bai
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Medicine, Tongji University, 200065 Shanghai, China
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine (BCM), Houston, Texas, 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas, 77030, USA
| | - Hua Wang
- Department of Medical Genetics, The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, (Hunan Children's Hospital), Changsha, Hunan 410007, China
| | - Shan Bian
- Institute for Regenerative Medicine, State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai 200120, China; National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, Hunan 410008, China; China Regional Research Center, International Center for Genetic Engineering and Biotechnology, Taizhou, Jiangsu 225316,China.
| | - Xiao Mao
- National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, University of South China, Changsha, Hunan 410008, China; Department of Medical Genetics, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, China.
| |
Collapse
|
27
|
Ahrentløv N, Kubrak O, Lassen M, Malita A, Koyama T, Frederiksen AS, Sigvardsen CM, John A, Madsen PEH, Halberg KV, Nagy S, Imig C, Richter EA, Texada MJ, Rewitz K. Protein-responsive gut hormone tachykinin directs food choice and impacts lifespan. Nat Metab 2025:10.1038/s42255-025-01267-0. [PMID: 40229448 DOI: 10.1038/s42255-025-01267-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/06/2025] [Indexed: 04/16/2025]
Abstract
Animals select food based on hungers that reflect dynamic macronutrient needs, but the hormonal mechanisms underlying nutrient-specific appetite regulation remain poorly defined. Here, we identify tachykinin (Tk) as a protein-responsive gut hormone in Drosophila and female mice, regulated by conserved environmental and nutrient-sensing mechanisms. Protein intake activates Tk-expressing enteroendocrine cells (EECs), driving the release of gut Tk through mechanisms involving target of rapamycin (TOR) and transient receptor potential A1 (TrpA1). In flies, we delineate a pathway by which gut Tk controls selective appetite and sleep after protein ingestion, mediated by glucagon-like adipokinetic hormone (AKH) signalling to neurons and adipose tissue. This mechanism suppresses protein appetite, promotes sugar hunger and modulates wakefulness to align behaviour with nutritional needs. Inhibiting protein-responsive gut Tk prolongs lifespan through AKH, revealing a role for nutrient-dependent gut hormone signalling in longevity. Our results provide a framework for understanding EEC-derived nutrient-specific satiety signals and the role of gut hormones in regulating food choice, sleep and lifespan.
Collapse
Affiliation(s)
- Nadja Ahrentløv
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Olga Kubrak
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mette Lassen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Alina Malita
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Takashi Koyama
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Amalie S Frederiksen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Casper M Sigvardsen
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Alphy John
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Kenneth V Halberg
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Stanislav Nagy
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Cordelia Imig
- Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark
- The Novo Nordisk Foundation, Hellerup, Denmark
| | - Erik A Richter
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Michael J Texada
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kim Rewitz
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
28
|
Mu X, Zhang Z, Liu Q, Ma J, Qin Y, Lang H, Zhang Y, Zhang N, Guo Q, Zhang P, Li D, Zhang R, Ji Q, Jiang A, Wang Y, Pan S, Liu X, Liu X, Sun J, Liu Y, Chen H, Zheng L, Meng L, Lu H, Zhang H, Zhai Y, Li Q, Liu J, Yang H, Wang J, Hu X, Xu X, Liu S, Zheng H. Single-nucleus and spatial transcriptomics identify brain landscape of gene regulatory networks associated with behavioral maturation in honeybees. Nat Commun 2025; 16:3343. [PMID: 40199930 PMCID: PMC11978848 DOI: 10.1038/s41467-025-58614-8] [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: 01/29/2023] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
Animal behavior is linked to the gene regulatory network (GRN) coordinating gene expression in the brain. Eusocial honeybees, with their natural behavioral plasticity, provide an excellent model for exploring the connection between brain activity and behavior. Using single-nucleus RNA sequencing and spatial transcriptomics, we analyze the expression patterns of brain cells associated with the behavioral maturation from nursing to foraging. Integrating spatial and cellular data uncovered cell-type and spatial heterogeneity in GRN organization. Interestingly, the stripe regulon is explicitly activated in foragers' small Keyon cells, which are implicated in spatial learning and navigation. When worker age is controlled in artificial colonies, stripe and its key targets remained highly expressed in the KC regions of bees performing foraging tasks. These findings suggest that specific GRNs coordinate individual brain cell activity during behavioral transitions, shedding light on GRN-driven brain heterogeneity and its role in the division of labor of social life.
Collapse
Affiliation(s)
- Xiaohuan Mu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Zijing Zhang
- College of Life Sciences, Hebei Normal University, Shijiazhuang, China
| | - Qun Liu
- BGI Research-Qingdao, BGI, Qingdao, China
| | - Jie Ma
- BGI Research-Qingdao, BGI, Qingdao, China
| | - Yating Qin
- BGI Research-Qingdao, BGI, Qingdao, China
| | - Haoyu Lang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | | | | | - Qunfei Guo
- BGI Research-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Pei Zhang
- BGI Research-Shenzhen, Shenzhen, China
| | - Denghui Li
- BGI Research-Qingdao, BGI, Qingdao, China
| | - Ruihua Zhang
- BGI Research-Qingdao, BGI, Qingdao, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Qianyue Ji
- BGI Research-Qingdao, BGI, Qingdao, China
| | | | - Yang Wang
- BGI Research-Shenzhen, Shenzhen, China
| | | | - Xiawei Liu
- BGI Research-Qingdao, BGI, Qingdao, China
| | - Xuemei Liu
- BGI Research-Qingdao, BGI, Qingdao, China
| | - Jiahui Sun
- BGI Research-Qingdao, BGI, Qingdao, China
| | - Yan Liu
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Hao Chen
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Li Zheng
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Liang Meng
- BGI Research-Qingdao, BGI, Qingdao, China
| | | | - He Zhang
- BGI Research-Shenzhen, Shenzhen, China
| | - Yifan Zhai
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
| | - Qiye Li
- BGI Research-Shenzhen, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | | | - Jian Wang
- BGI Research-Shenzhen, Shenzhen, China
| | - Xiaosong Hu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Xun Xu
- BGI Research-Shenzhen, Shenzhen, China.
| | | | - Hao Zheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.
| |
Collapse
|
29
|
Sivanantharajah L, Mudher A, Shepherd D. Examining the vulnerability of adult neuron subtypes to tau-mediated toxicity in Drosophila. Transl Psychiatry 2025; 15:127. [PMID: 40188067 PMCID: PMC11972385 DOI: 10.1038/s41398-025-03342-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Selective vulnerability of nerve cells is a feature of neurodegenerative disease. To date, animal models have been limited to examining pathogenic protein expression in broad or heterogeneous neuronal populations. Consequently, noted pathological hallmarks represent an average of disease phenotypes over multiple neuron types, rather than exact measures of individual responses. Here we targeted gene expression to small, precisely defined and homogenous neuronal populations in the Drosophila melanogaster central nervous system (CNS), allowing dissection of selective vulnerability of single types of neurons with single-neuron resolution. Using cellular degeneration as a readout for vulnerability, we found while all neurons were affected by tau some neuron types were more affected (vulnerable) than others (resilient). The tau-mediated pathogenic effects fell on a spectrum, demonstrating that neurons in the fly CNS are differentially vulnerable to tau pathology. Mechanistically, total tau levels did not correlate with vulnerability; rather, the best correlatives of degeneration were significant age-dependent increases in phospho-tau levels in the same neuron type, and tau mislocalisation into dendrites. Lastly, we found that tau phosphorylation in vulnerable neuron types correlated with downstream vesicular and mitochondrial trafficking defects. However, all vulnerable neuron types did not show the same pattern, suggesting multiple paths to degeneration. Beyond highlighting the heterogeneity of neuronal responses to tau in determining vulnerability, this work provides a new, high-resolution, tractable model for studying the age-dependent effects of tau, or any pathogenic protein, on postmitotic neurons with sub-cellular resolution.
Collapse
Affiliation(s)
| | - Amrit Mudher
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - David Shepherd
- Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
30
|
Pech U, Janssens J, Schoovaerts N, Kuenen S, Calatayud Aristoy C, Gallego SF, Makhzami S, Hulselmans GJ, Poovathingal S, Davie K, Bademosi AT, Swerts J, Vilain S, Aerts S, Verstreken P. Synaptic deregulation of cholinergic projection neurons causes olfactory dysfunction across five fly Parkinsonism models. eLife 2025; 13:RP98348. [PMID: 40178224 PMCID: PMC11968104 DOI: 10.7554/elife.98348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
The classical diagnosis of Parkinsonism is based on motor symptoms that are the consequence of nigrostriatal pathway dysfunction and reduced dopaminergic output. However, a decade prior to the emergence of motor issues, patients frequently experience non-motor symptoms, such as a reduced sense of smell (hyposmia). The cellular and molecular bases for these early defects remain enigmatic. To explore this, we developed a new collection of five fruit fly models of familial Parkinsonism and conducted single-cell RNA sequencing on young brains of these models. Interestingly, cholinergic projection neurons are the most vulnerable cells, and genes associated with presynaptic function are the most deregulated. Additional single nucleus sequencing of three specific brain regions of Parkinson's disease patients confirms these findings. Indeed, the disturbances lead to early synaptic dysfunction, notably affecting cholinergic olfactory projection neurons crucial for olfactory function in flies. Correcting these defects specifically in olfactory cholinergic interneurons in flies or inducing cholinergic signaling in Parkinson mutant human induced dopaminergic neurons in vitro using nicotine, both rescue age-dependent dopaminergic neuron decline. Hence, our research uncovers that one of the earliest indicators of disease in five different models of familial Parkinsonism is synaptic dysfunction in higher-order cholinergic projection neurons and this contributes to the development of hyposmia. Furthermore, the shared pathways of synaptic failure in these cholinergic neurons ultimately contribute to dopaminergic dysfunction later in life.
Collapse
Affiliation(s)
- Ulrike Pech
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Jasper Janssens
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Human Genetics, Leuven Brain InstituteLeuvenBelgium
- VIB-KU Leuven Center for AI and Computational Biology (VIB.AI)LeuvenBelgium
| | - Nils Schoovaerts
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Sabine Kuenen
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Carles Calatayud Aristoy
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Sandra F Gallego
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Samira Makhzami
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Human Genetics, Leuven Brain InstituteLeuvenBelgium
- VIB-KU Leuven Center for AI and Computational Biology (VIB.AI)LeuvenBelgium
| | - Gert J Hulselmans
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Human Genetics, Leuven Brain InstituteLeuvenBelgium
- VIB-KU Leuven Center for AI and Computational Biology (VIB.AI)LeuvenBelgium
| | - Suresh Poovathingal
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Human Genetics, Leuven Brain InstituteLeuvenBelgium
- VIB-KU Leuven Center for AI and Computational Biology (VIB.AI)LeuvenBelgium
- VIB-KU Leuven Center for Brain and Disease Research Technologies, Single Cell, Microfluidics and Bioinformatics Expertise UnitsLeuvenBelgium
| | - Kristofer Davie
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Human Genetics, Leuven Brain InstituteLeuvenBelgium
- VIB-KU Leuven Center for AI and Computational Biology (VIB.AI)LeuvenBelgium
- VIB-KU Leuven Center for Brain and Disease Research Technologies, Single Cell, Microfluidics and Bioinformatics Expertise UnitsLeuvenBelgium
| | - Adekunle T Bademosi
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Jef Swerts
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Sven Vilain
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| | - Stein Aerts
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Human Genetics, Leuven Brain InstituteLeuvenBelgium
- VIB-KU Leuven Center for AI and Computational Biology (VIB.AI)LeuvenBelgium
| | - Patrik Verstreken
- VIB-KU Leuven Center for Brain and Disease ResearchLeuvenBelgium
- KU Leuven, Department of Neurosciences, Leuven Brain InstituteLeuvenBelgium
| |
Collapse
|
31
|
Bereshneh AH, Andrews JC, Eberl DF, Bademci G, Borja NA, Bivona S, Chung WK, Yamamoto S, Wangler MF, McKee S, Tekin M, Bellen HJ, Kanca O. De novo variants in CDKL1 and CDKL2 are associated with neurodevelopmental symptoms. Am J Hum Genet 2025; 112:846-862. [PMID: 40088891 DOI: 10.1016/j.ajhg.2025.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025] Open
Abstract
The CDKL (cyclin-dependent kinase-like) family consists of five members in humans, CDKL1-5, that encode serine-threonine kinases. The only member that has been associated with a Mendelian disorder is CDKL5, and variants in CDKL5 cause developmental and epileptic encephalopathy type 2 (DEE2). Here, we study four de novo variants in CDKL2 identified in five individuals, including three unrelated probands and monozygotic twins. These individuals present with overlapping symptoms, including global developmental delay, intellectual disability, childhood-onset epilepsy, dyspraxia, and speech deficits. We also identified two individuals with de novo missense variants in CDKL1 in the published Deciphering Developmental Disorders (DDD) and GeneDx cohorts with developmental disorders. Drosophila has a single ortholog of CDKL1-5, CG7236 (Cdkl). Cdkl is expressed in sensory neurons that project to specific regions of the brain that control sensory inputs. Cdkl loss causes semi-lethality, climbing defects, heat-induced seizures, hearing loss, and reduced lifespan. These phenotypes can be rescued by expression of the human reference CDKL1, CDKL2, or CDKL5, showing that the functions of these genes are conserved. In contrast, the CDKL1 and CDKL2 variants do not fully rescue the observed phenotypes, and overexpression of the variant proteins leads to phenotypes that are similar to Cdkl loss. Co-expression of CDKL1 or CDKL2 variants with CDKL1, CDKL2, or CDKL5 references in the mutant background suppresses the rescue ability of the reference genes. Our results suggest that the variants act as dominant negative alleles and are causative of neurological symptoms in these individuals.
Collapse
Affiliation(s)
- Ali H Bereshneh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Jonathan C Andrews
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Daniel F Eberl
- Department of Biology, University of Iowa, Iowa City, IA, USA
| | - Guney Bademci
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Biomedical Research Building (BRB), Miami, FL, USA
| | - Nicholas A Borja
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Biomedical Research Building (BRB), Miami, FL, USA
| | - Stephanie Bivona
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Biomedical Research Building (BRB), Miami, FL, USA
| | - Wendy K Chung
- Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Michael F Wangler
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - Shane McKee
- Northern Ireland Regional Genetics Service, Belfast City Hospital, Belfast, Northern Ireland, UK
| | - Mustafa Tekin
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genetics, University of Miami Miller School of Medicine, Biomedical Research Building (BRB), Miami, FL, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA; Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA.
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
32
|
Chai C, Sultan E, Sarkar SR, Zhong L, Sarfati DN, Gershoni-Yahalom O, Jacobs-Wagner C, Rosental B, Wang B. Explosive cytotoxicity of 'ruptoblasts' bridges hormonal surveillance and immune defense. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645876. [PMID: 40236000 PMCID: PMC11996342 DOI: 10.1101/2025.03.28.645876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Cytotoxic killing is an essential immune function, yet its cellular mechanisms have been characterized in only a few model species. Here, we show that planarian flatworms harness a unique cytotoxic strategy. In planarians, activin, a hormone regulating regeneration and reproduction, also acts as an inflammatory cytokine. Overactivation of activin signaling - through protein injection, genetic chimerism, or bacterial infection - triggers 'ruptoblasts', an undocumented immune cell type, to undergo 'ruptosis', a unique mode of cell bursting that eliminates nearby cells and bacteria in mere minutes, representing one of the fastest cytotoxic mechanisms observed. Ablating ruptoblasts suppresses inflammation but compromises bacterial clearance, highlighting ruptoblasts' broad-spectrum immune functions. We further identified ruptoblast-like cells in diverse basal bilaterians, unveiling an alternative strategy that couples hormonal regulation with immune defense and expanding the landscape of evolutionary immune innovations.
Collapse
|
33
|
Brunet M, Thomas J, Lapart JA, Krüttli L, Laporte MH, Riparbelli MG, Callaini G, Durand B, Morel V. Drosophila Alms1 proteins regulate centriolar cartwheel assembly by enabling Plk4-Ana2 amplification loop. EMBO J 2025; 44:2366-2395. [PMID: 40021845 PMCID: PMC12000580 DOI: 10.1038/s44318-025-00382-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 01/06/2025] [Accepted: 01/24/2025] [Indexed: 03/03/2025] Open
Abstract
Centrioles play a central role in cell division by recruiting pericentriolar material (PCM) to form the centrosome. Alterations in centriole number or function lead to various diseases including cancer or microcephaly. Centriole duplication is a highly conserved mechanism in eukaryotes. Here, we show that the two Drosophila orthologs of the Alström syndrome protein 1 (Alms1a and Alms1b) are unexpected novel players of centriole duplication in fly. Using Ultrastructure Expansion Microscopy, we reveal that Alms1a is a PCM protein that is loaded proximally on centrioles at the onset of procentriole formation, whereas Alms1b caps the base of mature centrioles. We demonstrate that chronic loss of Alms1 proteins (with RNA null alleles) affects PCM maturation, whereas their acute loss (in RNAi KD) completely disrupts procentriole formation before Sas-6 cartwheel assembly. We establish that Alms1 proteins are required for the amplification of the Plk4-Ana2 pool at the duplication site and the subsequent Sas-6 recruitment. Thus, Alms1 proteins are novel critical but highly buffered regulators of PCM and cartwheel assembly in flies.
Collapse
Affiliation(s)
- Marine Brunet
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Joëlle Thomas
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Jean-André Lapart
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Léo Krüttli
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | - Marine H Laporte
- Universite Claude BERNARD Lyon 1, Lyon, France
- MeLiS-CNRS-UMR5284, Lyon, France
- INSERM-U1314, Lyon, France
| | | | | | - Bénédicte Durand
- Universite Claude BERNARD Lyon 1, Lyon, France.
- MeLiS-CNRS-UMR5284, Lyon, France.
- INSERM-U1314, Lyon, France.
| | - Véronique Morel
- Universite Claude BERNARD Lyon 1, Lyon, France.
- MeLiS-CNRS-UMR5284, Lyon, France.
- INSERM-U1314, Lyon, France.
| |
Collapse
|
34
|
Nässel DR. What Drosophila can tell us about state-dependent peptidergic signaling in insects. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2025; 179:104275. [PMID: 39956367 DOI: 10.1016/j.ibmb.2025.104275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Plasticity in animal behavior and physiology is largely due to modulatory and regulatory signaling with neuropeptides and peptide hormones (collectively abbreviated NPHs). The NPHs constitute a very large and versatile group of signaling substances that partake at different regulatory levels in most daily activities of an organism. This review summarizes key principles in NPH actions in the brain and in interorgan signaling, with focus on Drosophila. NPHs are produced by neurons, neurosecretory cells (NSCs) and other endocrine cells in NPH-specific and stereotypic patterns. Most of the NPHs have multiple (pleiotropic) functions and target several different neuronal circuits and/or peripheral tissues. Such divergent NPH signaling ensures orchestration of behavior and physiology in state-dependent manners. Conversely, many neurons, circuits, NSCs, or other cells, are targeted by multiple NPHs. This convergent signaling commonly conveys various signals reporting changes in the external and internal environment to central neurons/circuits. As an example of wider functional convergence, 26 different Drosophila NPHs act at many different levels to regulate food search and feeding. Convergence is also seen in hormonal regulation of peripheral functions. For instance, multiple NPHs target renal tubules to ensure osmotic homeostasis. Interestingly, several of the same osmoregulatory NPHs also regulate feeding, metabolism and stress. However, for some NPHs the cellular distribution and functions suggests multiple unrelated functions that are restricted to specific circuits. Thus, NPH signaling follows distinct patterns for each specific NPH, but taken together they form overlapping networks that modulate behavior and physiology.
Collapse
Affiliation(s)
- Dick R Nässel
- Department of Zoology, Stockholm University, S-10691, Stockholm, Sweden.
| |
Collapse
|
35
|
Lee D, Shahandeh MP, Abuin L, Benton R. Comparative single-cell transcriptomic atlases of drosophilid brains suggest glial evolution during ecological adaptation. PLoS Biol 2025; 23:e3003120. [PMID: 40299832 PMCID: PMC12040179 DOI: 10.1371/journal.pbio.3003120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 03/17/2025] [Indexed: 05/01/2025] Open
Abstract
To explore how brains change upon species evolution, we generated single-cell transcriptomic atlases of the central brains of three closely related but ecologically distinct drosophilids: the generalists Drosophila melanogaster and Drosophila simulans, and the noni fruit specialist Drosophila sechellia. The global cellular composition of these species' brains is well-conserved, but we predicted a few cell types with different frequencies, notably perineurial glia of the blood-brain barrier, which we validate in vivo. Gene expression analysis revealed that distinct cell types evolve at different rates and patterns, with glial populations exhibiting the greatest divergence between species. Compared to the D. melanogaster brain, cellular composition and gene expression patterns are more divergent in D. sechellia than in D. simulans-despite their similar phylogenetic distance from D. melanogaster-indicating that the specialization of D. sechellia is reflected in the structure and function of its brain. Expression changes in D. sechellia include several metabolic signaling genes, suggestive of adaptations to its novel source of nutrition. Additional single-cell transcriptomic analysis on D. sechellia revealed genes and cell types responsive to dietary supplement with noni, pointing to glia as sites for both physiological and genetic adaptation to this fruit. Our atlases represent the first comparative datasets for "whole" central brains and provide a comprehensive foundation for studying the evolvability of nervous systems in a well-defined phylogenetic and ecological framework.
Collapse
Affiliation(s)
- Daehan Lee
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Biological Sciences, College of Natural Sciences, Sungkyunkwan University, Suwon, Republic of Korea
| | - Michael P. Shahandeh
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Biology, Hofstra University, Hempstead, New York, United States of America
| | - Liliane Abuin
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Richard Benton
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
36
|
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
|
37
|
Zhang W, Nie Y, Xu T, Li Y, Xu Y, Chen X, Shi P, Liu F, Zhao H, Ma Q, Xu J. Evolutionary Process Underlying Receptor Gene Expansion and Cellular Divergence of Olfactory Sensory Neurons in Honeybees. Mol Biol Evol 2025; 42:msaf080. [PMID: 40172919 PMCID: PMC12001030 DOI: 10.1093/molbev/msaf080] [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: 01/02/2025] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025] Open
Abstract
Olfaction is crucial for animals' survival and adaptation. Unlike the strict singular expression of odorant receptor (OR) genes in vertebrate olfactory sensory neurons (OSNs), insects exhibit complex OR gene expression patterns. In honeybees (Apis mellifera), a significant expansion of OR genes implies a selection preference for the olfactory demands of social insects. However, the mechanisms underlying receptor expression specificity and their contribution to OSN divergence remain unclear. In this study, we used single-nucleus multiomics profiling to investigate the transcriptional regulation of OR genes and the cellular identity of OSNs in A. mellifera. We identified three distinct OR expression patterns, singular OR expression, co-expression of multiple OR genes with a single active promoter, and co-expression of multiple OR genes with multiple active promoters. Notably, ∼50% of OSNs co-expressed multiple OR genes, driven by polycistronic transcription of tandemly duplicated OR genes via a single active promoter. In these OSNs, their identity was determined by the first transcribed receptor. The divergent activation of the promoter for duplicated OR genes ensures the coordinated increased divergence of OSN population. By integrating multiomics data with genomic architecture, we illustrate how fundamental genetic mechanisms drive OR gene expansion and influence flanking regulatory elements, ultimately contributing to the cellular divergence of OSNs. Our findings highlight the interplay between gene duplication and regulatory evolution in shaping OSN diversity, providing new insights into the evolution and adaptation of olfaction in social insects. This study also sheds light on how genetic innovations contribute to the evolution of complex traits.
Collapse
Affiliation(s)
- Weixing Zhang
- State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yage Nie
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Tao Xu
- State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yiheng Li
- State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Yicong Xu
- State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Xiaoyong Chen
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510080, China
| | - Peiyu Shi
- State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| | - Fang Liu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510000, China
| | - Hongxia Zhao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou 510000, China
| | - Qing Ma
- Center for Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jin Xu
- State Key Laboratory of Biocontrol, Innovation Center for Evolutionary Synthetic Biology, School of Life Sciences, Sun Yat-Sen University, Guangzhou 510275, China
| |
Collapse
|
38
|
Oka M, Nakajima S, Suzuki E, Yamamoto S, Ando K. Glucose uptake in pigment glia suppresses Tau-induced inflammation and photoreceptor degeneration. Dis Model Mech 2025; 18:dmm052057. [PMID: 40151148 PMCID: PMC12067088 DOI: 10.1242/dmm.052057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Brain inflammation contributes to the pathogenesis of neurodegenerative diseases, such as Alzheimer's disease (AD). Glucose hypometabolism and glial activation are pathological features seen in AD brains; however, the connection between the two is not fully understood. Using a Drosophila model of AD, we identified that glucose metabolism in glia plays a critical role in neuroinflammation under disease conditions. Expression of human MATP (hereafter referred to as Tau) in the retinal cells, including photoreceptor neurons and pigment glia, causes photoreceptor degeneration accompanied by the formation of dark-stained round inclusion-like structures and swelling of the lamina cortex. We found that inclusion-like structures are formed by glial phagocytosis, and swelling of the laminal cortex correlates with the expression of antimicrobial peptides. Coexpression of human glucose transporter 3 (SLC2A3, hereafter referred to as GLUT3) with Tau in the retina does not affect Tau levels but suppresses these inflammatory responses and photoreceptor degeneration. We also found that expression of GLUT3, specifically in the pigment glia, is sufficient to suppress inflammatory phenotypes and mitigate photoreceptor degeneration in the Tau-expressing retina. Our results suggest that glial glucose metabolism contributes to inflammatory responses and neurodegeneration in tauopathy.
Collapse
Affiliation(s)
- Mikiko Oka
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute for Texas Children's Hospital, Houston, TX 77030, USA
| | - Sho Nakajima
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Emiko Suzuki
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Jan and Dan Duncan Neurological Research Institute for Texas Children's Hospital, Houston, TX 77030, USA
| | - Kanae Ando
- Department of Biological Sciences, Tokyo Metropolitan University, Tokyo 192-0397, Japan
| |
Collapse
|
39
|
Simmons C, Williams IH, Bradshaw TW, Armstrong AR. Adipocyte-Derived CCHamide-1, Eiger, Growth-Blocking Peptide 3, and Unpaired 2 Regulate Drosophila melanogaster Oogenesis. Biomolecules 2025; 15:513. [PMID: 40305230 PMCID: PMC12024527 DOI: 10.3390/biom15040513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
In addition to energy storage, adipose tissue communication to other organs plays a key role in regulating organismal physiology. While the link between adipose tissue dysfunction and pathophysiology, including diabetes, chronic inflammation, and infertility, is clear, the molecular mechanisms that underlie these associations have not been fully described. We use Drosophila melanogaster as a model to better understand how adipose tissue communicates to the ovary. In this study, we utilized D. melanogaster's robust genetic toolkit to examine the role of five adipokines known to control larval growth during development, CCHamide-1, CCHamide-2, eiger, Growth-blocking peptide 3, and unpaired 2 in regulating oogenesis. We show that the adult fat body expresses these "larval" adipokines. Our data indicate that ovarian germline stem cell maintenance does not require these adipokines. However, adipocyte-derived CCHamide-1, eiger, Growth-blocking peptide 3, and unpaired 2 influence early and late germline survival as well as ovulation. Thus, this work uncovers several adipokines that mediate fat-to-ovary communication.
Collapse
Affiliation(s)
| | | | | | - Alissa Richmond Armstrong
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC 29208, USA; (C.S.); (I.H.W.); (T.W.B.)
| |
Collapse
|
40
|
Puig-Barbe A, Dettmann S, Nirello VD, Moor H, Azami S, Edgar BA, Varga-Weisz P, Korzelius J, de Navascués J. A bHLH interaction code controls bipotential differentiation and self-renewal in the Drosophila gut. Cell Rep 2025; 44:115398. [PMID: 40089983 DOI: 10.1016/j.celrep.2025.115398] [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: 09/19/2023] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 03/18/2025] Open
Abstract
Multipotent adult stem cells balance self-renewal with differentiation into various cell types. How this balance is regulated at the transcriptional level is poorly understood. Here, we show that a network of basic helix-loop-helix (bHLH) transcription factors controls both stemness and bipotential differentiation in the Drosophila adult intestine. We find that homodimers of Daughterless (Da), a homolog of mammalian E proteins, maintain self-renewal of intestinal stem cells (ISCs), antagonizing the enteroendocrine fate promoted by heterodimers of Da and Scute (Sc; homolog of ASCL). The HLH factor Extramacrochaetae (Emc; homologous to Id proteins) promotes absorptive differentiation by titrating Da and Sc. Emc prevents the committed absorptive progenitor from dedifferentiating, underscoring the plasticity of these cells. Switching physical interaction partners in this way enables the active maintenance of stemness while priming stem cells for differentiation along two alternative fates. Such regulatory logic is likely operative in other bipotent stem cell systems.
Collapse
Affiliation(s)
- Aleix Puig-Barbe
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK
| | - Svenja Dettmann
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; AbbVie Germany GmbH & Co. KG, 81 Mainzer Str., 65189 Wiesbaden, Frankfurt, Germany
| | - Vinícius Dias Nirello
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil
| | - Helen Moor
- School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Sina Azami
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Straße 9B, 50931 Köln, Germany
| | - Bruce A Edgar
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Huntsman Cancer Institute & Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
| | - Patrick Varga-Weisz
- International Laboratory for Microbiome Host Epigenetics, Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas, SP 13083-862, Brazil; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK
| | - Jerome Korzelius
- DKFZ/ZMBH Alliance, University of Heidelberg, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Straße 9B, 50931 Köln, Germany; School of Biosciences, University of Kent, Canterbury CT2 7NZ, UK
| | - Joaquín de Navascués
- School of Biosciences, Cardiff University, The Sir Martin Evans Building, Museum Avenue, Cardiff CF10 3AX, UK; School of Life Sciences, University of Essex, Wivenhoe Park, Colchester CO4 3SQ, UK.
| |
Collapse
|
41
|
Janssens J, Mangeol P, Hecker N, Partel G, Spanier KI, Ismail JN, Hulselmans GJ, Aerts S, Schnorrer F. Spatial transcriptomics in the adult Drosophila brain and body. eLife 2025; 13:RP92618. [PMID: 40100257 PMCID: PMC11919255 DOI: 10.7554/elife.92618] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Recently, we have achieved a significant milestone with the creation of the Fly Cell Atlas. This single-nuclei atlas encompasses the entire fly, covering the entire head and body, in addition to all major organs. This atlas catalogs many hundreds of cell types, of which we annotated 250. Thus, a large number of clusters remain to be fully characterized, in particular in the brain. Furthermore, by applying single-nuclei sequencing, all information about the spatial location of the cells in the body and of about possible subcellular localization of the mRNAs within these cells is lost. Spatial transcriptomics promises to tackle these issues. In a proof-of-concept study, we have here applied spatial transcriptomics using a selected gene panel to pinpoint the locations of 150 mRNA species in the adult fly. This enabled us to map unknown clusters identified in the Fly Cell Atlas to their spatial locations in the fly brain. Additionally, spatial transcriptomics discovered interesting principles of mRNA localization and transcriptional diversity within the large and crowded muscle cells that may spark future mechanistic investigations. Furthermore, we present a set of computational tools that will allow for easier integration of spatial transcriptomics and single-cell datasets.
Collapse
Affiliation(s)
- Jasper Janssens
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Pierre Mangeol
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| | - Nikolai Hecker
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Gabriele Partel
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Katina I Spanier
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Joy N Ismail
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
| | - Gert J Hulselmans
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Stein Aerts
- VIB-KU Leuven Center for Brain and Disease Research, KU LeuvenLeuvenBelgium
- Laboratory of Computational Biology, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for AI & Computational Biology, KU LeuvenLeuvenBelgium
| | - Frank Schnorrer
- Aix Marseille University, CNRS, IBDM, Turing Centre for Living SystemsMarseilleFrance
| |
Collapse
|
42
|
Garrido VE, Outlaw WG, Powell AM, Ables ET. SR Protein Kinase is expressed in Drosophila ovarian germline stem cells but is not essential for their self-renewal. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001550. [PMID: 40171240 PMCID: PMC11959418 DOI: 10.17912/micropub.biology.001550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/07/2025] [Accepted: 03/15/2025] [Indexed: 04/03/2025]
Abstract
Germline stem cells (GSCs) are necessary for oocyte production in Drosophila . GSC maintenance is regulated by intrinsic factors that promote their self-renewal. One such factor, the beta-importin, Transportin-Serine/Arginine rich , mediates nuclear import of serine/arginine-rich (SR) proteins, which are phosphorylated by SR protein kinases. Here, we investigate whether the kinase encoded by SR protein kinase ( SRPK ) is essential for GSC self-renewal. We find that SRPK is expressed in GSCs and their mitotically-dividing daughters, but is not necessary for GSC establishment or maintenance. We conclude that SRPK is dispensable for GSC self-renewal, and postulate that other protein kinases can compensate for its absence.
Collapse
Affiliation(s)
- Victoria E. Garrido
- Biology, East Carolina University, Greenville, North Carolina, United States
| | - William G. Outlaw
- Biology, East Carolina University, Greenville, North Carolina, United States
| | - Amanda M. Powell
- Biology, East Carolina University, Greenville, North Carolina, United States
| | - Elizabeth T. Ables
- Biology, East Carolina University, Greenville, North Carolina, United States
| |
Collapse
|
43
|
Akagi K, Jin YJ, Koizumi K, Oku M, Ito K, Shen X, Imura JI, Aihara K, Saito S. Integration of Dynamical Network Biomarkers, Control Theory and Drosophila Model Identifies Vasa/DDX4 as the Potential Therapeutic Targets for Metabolic Syndrome. Cells 2025; 14:415. [PMID: 40136664 PMCID: PMC11941168 DOI: 10.3390/cells14060415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/05/2025] [Accepted: 03/09/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic syndrome (MetS) is a subclinical disease, resulting in increased risk of type 2 diabetes (T2D), cardiovascular diseases, cancer, and mortality. Dynamical network biomarkers (DNB) theory has been developed to provide early-warning signals of the disease state during a preclinical stage. To improve the efficiency of DNB analysis for the target genes discovery, the DNB intervention analysis based on the control theory has been proposed. However, its biological validation in a specific disease such as MetS remains unexplored. Herein, we identified eight candidate genes from adipose tissue of MetS model mice at the preclinical stage by the DNB intervention analysis. Using Drosophila, we conducted RNAi-mediated knockdown screening of these candidate genes and identified vasa (also known as DDX4), encoding a DEAD-box RNA helicase, as a fat metabolism-associated gene. Fat body-specific knockdown of vasa abrogated high-fat diet (HFD)-induced enhancement of starvation resistance through up-regulation of triglyceride lipase. We also confirmed that DDX4 expressing adipocytes are increased in HFD-fed mice and high BMI patients using the public datasets. These results prove the potential of the DNB intervention analysis to search the therapeutic targets for diseases at the preclinical stage.
Collapse
Affiliation(s)
- Kazutaka Akagi
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan;
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Ying-Jie Jin
- Graduate School of Pharma-Medical Sciences, University of Toyama, Toyama 930-0194, Japan;
| | - Keiichi Koizumi
- Division of Presymptomatic Disease, Institute of Natural Medicine, University of Toyama, Toyama 930-0194, Japan;
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Makito Oku
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| | - Kaisei Ito
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan;
| | - Xun Shen
- Graduate School of Bio-Applications and Systems Engineering, Tokyo University of Agriculture and Technology, Tokyo 184-8588, Japan;
| | - Jun-ichi Imura
- Department of Systems and Control Engineering, School of Engineering, Institute of Science Tokyo, Tokyo 152-8552, Japan;
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo, Tokyo 113-0033, Japan;
| | - Shigeru Saito
- Research Center for Pre-Disease Science, University of Toyama, Toyama 930-8555, Japan; (M.O.); (S.S.)
| |
Collapse
|
44
|
Moore J, Wu T, Dhindsa J, El Fadel O, Le A, Perez A, Amoh B, Tarkunde A, Zhu KF, Avalos M, Dammer EB, Duong DM, Seyfried NT, Shulman JM, Al-Ramahi I, Botas J. Longitudinal multi-omics in alpha-synuclein Drosophila model discriminates disease- from age-associated pathologies in Parkinson's disease. NPJ Parkinsons Dis 2025; 11:46. [PMID: 40069190 PMCID: PMC11897226 DOI: 10.1038/s41531-025-00899-z] [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] [Received: 08/15/2024] [Accepted: 02/17/2025] [Indexed: 03/15/2025] Open
Abstract
Parkinson's disease (PD) starts decades before symptoms appear, usually in the later decades of life, when age-related changes are occurring. To identify molecular changes early in the disease course and distinguish PD pathologies from aging, we generated Drosophila expressing alpha-synuclein (αSyn) in neurons and performed longitudinal bulk transcriptomics and proteomics on brains at six time points across the lifespan and compared the data to healthy control flies as well as human post-mortem brain datasets. We found that translational and energy metabolism pathways were downregulated in αSyn flies at the earliest timepoints; comparison with the aged control flies suggests that elevated αSyn accelerates changes associated with normal aging. Unexpectedly, single-cell analysis at a mid-disease stage revealed that neurons upregulate protein synthesis and nonsense-mediated decay, while glia drive their overall downregulation. Longitudinal multi-omics approaches in animal models can thus help elucidate the molecular cascades underlying neurodegeneration vs. aging and co-pathologies.
Collapse
Affiliation(s)
- Justin Moore
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Quantitative and Computational Bioscience Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Timothy Wu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Justin Dhindsa
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Omar El Fadel
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Anh Le
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Alma Perez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Bismark Amoh
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Akash Tarkunde
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Katy F Zhu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Matthew Avalos
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Duc M Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Joshua M Shulman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Deparment of Neurology, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ismael Al-Ramahi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA.
- Quantitative and Computational Bioscience Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, 77030, USA.
- Center for Alzheimer's and Neurodegenerative Diseases, Baylor College of Medicine, Houston, TX, 77030, USA.
- Genetics and Genomics Graduate Program, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
45
|
Held M, Bisen RS, Zandawala M, Chockley AS, Balles IS, Hilpert S, Liessem S, Cascino-Milani F, Ache JM. Aminergic and peptidergic modulation of insulin-producing cells in Drosophila. eLife 2025; 13:RP99548. [PMID: 40063677 PMCID: PMC11893105 DOI: 10.7554/elife.99548] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/13/2025] Open
Abstract
Insulin plays a critical role in maintaining metabolic homeostasis. Since metabolic demands are highly dynamic, insulin release needs to be constantly adjusted. These adjustments are mediated by different pathways, most prominently the blood glucose level, but also by feedforward signals from motor circuits and different neuromodulatory systems. Here, we analyze how neuromodulatory inputs control the activity of the main source of insulin in Drosophila - a population of insulin-producing cells (IPCs) located in the brain. IPCs are functionally analogous to mammalian pancreatic beta cells, but their location makes them accessible for in vivo recordings in intact animals. We characterized functional inputs to IPCs using single-nucleus RNA sequencing analysis, anatomical receptor expression mapping, connectomics, and an optogenetics-based 'intrinsic pharmacology' approach. Our results show that the IPC population expresses a variety of receptors for neuromodulators and classical neurotransmitters. Interestingly, IPCs exhibit heterogeneous receptor profiles, suggesting that the IPC population can be modulated differentially. This is supported by electrophysiological recordings from IPCs, which we performed while activating different populations of modulatory neurons. Our analysis revealed that some modulatory inputs have heterogeneous effects on the IPC activity, such that they inhibit one subset of IPCs, while exciting another. Monitoring calcium activity across the IPC population uncovered that these heterogeneous responses occur simultaneously. Certain neuromodulatory populations shifted the IPC population activity towards an excited state, while others shifted it towards inhibition. Taken together, we provide a comprehensive, multi-level analysis of neuromodulation in the insulinergic system of Drosophila.
Collapse
Affiliation(s)
- Martina Held
- Ache Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| | - Rituja S Bisen
- Ache Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| | - Meet Zandawala
- Zandawala Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
- Department of Biochemistry and Molecular Biology, University of Nevada RenoRenoUnited States
| | - Alexander S Chockley
- Ache Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| | - Isabella S Balles
- Ache Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| | - Selina Hilpert
- Zandawala Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| | - Sander Liessem
- Ache Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| | - Federico Cascino-Milani
- Ache Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| | - Jan M Ache
- Ache Lab, Neurobiology and Genetics, Theodor-Boveri-Institute, Biocenter, Julius-Maximilians-University of Würzburg, Am HublandWürzburgGermany
| |
Collapse
|
46
|
Rust K, Schubert A, Peralta JM, Nystul TG. Independent signaling pathways provide a fail-safe mechanism to prevent tumorigenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640798. [PMID: 40093137 PMCID: PMC11908167 DOI: 10.1101/2025.02.28.640798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Controlled signaling activity is vital for normal tissue homeostasis and oncogenic signaling activation facilitates tumorigenesis. Here we use single-cell transcriptomics to investigate the effects of pro-proliferative signaling on epithelial homeostasis using the Drosophila follicle cell lineage. Notably, EGFR-Ras overactivation induces cell cycle defects by activating the transcription factors Pointed and E2f1 and impedes differentiation. Hh signaling simultaneously promotes an undifferentiated state and induces differentiation via activation of EMT-associated transcription factors zfh1 and Mef2. As a result, overactivation of Hh signaling generates a transcriptional hybrid state comparable to epithelial-mesenchymal-transition. Co-overactivation of Hh signaling with EGFR-Ras signaling blocks differentiation and induces key characteristics of tumor cells including a loss of tissue architecture caused by reduced expression of cell adhesion molecules, sustained proliferation and an evasion of cell cycle checkpoints. These findings provide new insight into how non-interacting signaling pathways converge at the transcriptional level to prevent malignant cell behavior.
Collapse
Affiliation(s)
- Katja Rust
- Institute of Physiology and Pathophysiology, Dept. of Molecular Cell Physiology, Philipps University Marburg, Germany
| | - Andrea Schubert
- Institute of Physiology and Pathophysiology, Dept. of Molecular Cell Physiology, Philipps University Marburg, Germany
| | - Jobelle M Peralta
- UCSF, Department of Anatomy, 513 Parnassus Ave, San Francisco, CA 94143, USA
- UCSF, Department of OB-GYN/RS, 513 Parnassus Ave, San Francisco, CA 94143, USA
- Broad Center of Regeneration Medicine and Stem Cell Research, 513 Parnassus Ave, San Francisco, CA 94143, USA
| | - Todd G Nystul
- UCSF, Department of Anatomy, 513 Parnassus Ave, San Francisco, CA 94143, USA
- UCSF, Department of OB-GYN/RS, 513 Parnassus Ave, San Francisco, CA 94143, USA
- Broad Center of Regeneration Medicine and Stem Cell Research, 513 Parnassus Ave, San Francisco, CA 94143, USA
| |
Collapse
|
47
|
Epiney D, Morales Chaya GN, Dillon NR, Lai SL, Doe CQ. Transcriptional complexity in the insect central complex: single nuclei RNA-sequencing of adult brain neurons derived from type 2 neuroblasts. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.12.10.571022. [PMID: 40093129 PMCID: PMC11908175 DOI: 10.1101/2023.12.10.571022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
In both invertebrates such as Drosophila and vertebrates such as mouse or human, the brain contains the most diverse population of cell types of any tissue. It is generally accepted that transcriptional diversity is an early step in generating neuronal and glial diversity, followed by the establishment of a unique gene expression profile that determines morphology, connectivity, and function. In Drosophila, there are two types of neural stem cells, called Type 1 (T1) and Type 2 (T2) neuroblasts. In contrast to T1 neuroblasts, T2 neuroblasts generate intermediate neural progenitors (INPs) that expand the number and diversity of cell types. The diversity of T2-derived neurons contributes a large portion of the central complex (CX), a conserved brain region that plays a role in sensorimotor integration. Recent work has revealed much of the connectome of the CX, but how this connectome is assembled remains unclear. Mapping the transcriptional diversity of neurons derived from T2 neuroblasts is a necessary step in linking transcriptional profile to the assembly of the adult brain. Here we perform single nuclei RNA sequencing of T2 neuroblast-derived adult neurons and glia. We identify clusters containing all known classes of glia, clusters that are male/female enriched, and 161 neuron-specific clusters. We map neurotransmitter and neuropeptide expression and identify unique transcription factor combinatorial codes for each cluster (presumptive neuron subtype). This is a necessary step that directs functional studies to determine whether each transcription factor combinatorial code specifies a distinct neuron type within the CX. We map several columnar neuron subtypes to distinct clusters and identify two neuronal classes (NPF+ and AstA+) that both map to two closely related clusters. Our data support the hypothesis that each transcriptional cluster represents one or a few closely related neuron subtypes.
Collapse
Affiliation(s)
| | | | | | - Sen-Lin Lai
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| | - Chris Q. Doe
- Institute of Neuroscience, Howard Hughes Medical Institute, University of Oregon, Eugene, OR 97403
| |
Collapse
|
48
|
Purice MD, Lago‐Baldaia I, Fernandes VM, Singhvi A. Molecular profiling of invertebrate glia. Glia 2025; 73:632-656. [PMID: 39415317 PMCID: PMC11784859 DOI: 10.1002/glia.24623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/06/2024] [Accepted: 09/18/2024] [Indexed: 10/18/2024]
Abstract
Caenorhabditis elegans and Drosophila melanogaster are powerful experimental models for uncovering fundamental tenets of nervous system organization and function. Findings over the last two decades show that molecular and cellular features are broadly conserved between invertebrates and vertebrates, indicating that insights derived from invertebrate models can broadly inform our understanding of glial operating principles across diverse species. In recent years, these model systems have led to exciting discoveries in glial biology and mechanisms of glia-neuron interactions. Here, we summarize studies that have applied current state-of-the-art "-omics" techniques to C. elegans and D. melanogaster glia. Coupled with the remarkable acceleration in the pace of mechanistic studies of glia biology in recent years, these indicate that invertebrate glia also exhibit striking molecular complexity, specificity, and heterogeneity. We provide an overview of these studies and discuss their implications as well as emerging questions where C. elegans and D. melanogaster are well-poised to fill critical knowledge gaps in our understanding of glial biology.
Collapse
Affiliation(s)
- Maria D. Purice
- Division of Basic SciencesFred Hutchinson Cancer CenterSeattleWashingtonUSA
- Department of Biological StructureSchool of Medicine, University of WashingtonSeattleWashingtonUSA
| | - Inês Lago‐Baldaia
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | | | - Aakanksha Singhvi
- Division of Basic SciencesFred Hutchinson Cancer CenterSeattleWashingtonUSA
- Department of Biological StructureSchool of Medicine, University of WashingtonSeattleWashingtonUSA
| |
Collapse
|
49
|
Sun Z, Li L, Zhang L. Apigenin enhancing oxidative resistance and proteostasis to extend lifespan via PTEN-mediated AKT signalling pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167670. [PMID: 39826849 DOI: 10.1016/j.bbadis.2025.167670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/27/2024] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
Aging is a complicated process, featuring the progressive deterioration of physiological functions and a heightened susceptibility to diseases including neurodegenerative disorders, cardiovascular diseases, and cancer. Apigenin, a flavonoid existing in various plants, has attracted attention due to its potential role in anti-aging. In this investigation, the potential effect of apigenin on extending lifespan in Saccharomyces cerevisiae (yeast) and Drosophila melanogaster (flies) was explored. The results indicate that apigenin significantly extends both replicative and chronological life duration in yeast, as well as longevity in male and female flies. Apigenin treatment also improves resistance to oxidative stress in both organisms, as manifested by enhanced survival, decreased reactive oxygen species (ROS) levels and upregulation of antioxidant enzymes. Furthermore, apigenin activates crucial elements of the proteostasis network (PN), such as upregulation of proteostasis-related enzymes activity and genes expression. Network analysis revealed that apigenin affects aging conserved in the longevity-regulating pathway. Notably, Pten is a hub target in flies. Apigenin regulated DmPten at both mRNA and protein expression level while modulating downstream targets, including the phosphorylation of AKT and associated signalling pathways. In a high-sucrose diet (HSD) model, Apigenin treatment extended lifespan, reduced hemolymph glucose levels, enhanced Pten expression, suppressed AKT phosphorylation, and modulated the phosphorylation status of S6K and expression of DmFoxo. These results demonstrate that apigenin could serve as a longevity research object and potential therapeutic drug for promoting health and longevity through its antioxidant and proteostatic properties.
Collapse
Affiliation(s)
- Zhengqiong Sun
- State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Hangzhou 311300, China
| | - Lei Li
- Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China
| | - Lei Zhang
- State Key Laboratory of Subtropical Silviculture, Zhejiang A & F University, Hangzhou 311300, China; Department of Pharmaceutical Botany, School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
50
|
Briney CA, Henriksen JC, Lin C, Jones LA, Benner L, Rains AB, Gutierrez R, Gafken PR, Rissland OS. Muskelin is a substrate adaptor of the highly regulated Drosophila embryonic CTLH E3 ligase. EMBO Rep 2025; 26:1647-1669. [PMID: 39979464 PMCID: PMC11933467 DOI: 10.1038/s44319-025-00397-6] [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: 07/24/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
The maternal-to-zygotic transition (MZT) is a conserved developmental process where the maternally-derived protein and mRNA cache is replaced with newly made zygotic gene products. We have previously shown that in Drosophila the deposited RNA-binding proteins ME31B, Cup, and Trailer Hitch are ubiquitylated by the CTLH E3 ligase and cleared. However, the organization and regulation of the CTLH complex remain poorly understood in flies because Drosophila lacks an identifiable substrate adaptor, and the mechanisms restricting the degradation of ME31B and its cofactors to the MZT are unknown. Here, we show that the developmental regulation of the CTLH complex is multi-pronged, including transcriptional control by OVO and autoinhibition of the E3 ligase. One major regulatory target is the subunit Muskelin, which we demonstrate is a substrate adaptor for the Drosophila CTLH complex. Finally, we find that Muskelin has few targets beyond the three known RNA-binding proteins, showing exquisite target specificity. Thus, multiple levels of integrated regulation restrict the activity of the embryonic CTLH complex to early embryogenesis, during which time it regulates three important RNA-binding proteins.
Collapse
Affiliation(s)
- Chloe A Briney
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Jesslyn C Henriksen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Chenwei Lin
- Proteomics & Metabolomics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Lisa A Jones
- Proteomics & Metabolomics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Leif Benner
- Section of Developmental Genomics, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Addison B Rains
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Roxana Gutierrez
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Philip R Gafken
- Proteomics & Metabolomics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Olivia S Rissland
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
- RNA Bioscience Initiative, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|