1
|
Sears JC, Broadie K. PKA restricts ERK signaling in learning and memory Kenyon cell neurons. Cell Signal 2025; 132:111818. [PMID: 40250698 DOI: 10.1016/j.cellsig.2025.111818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Protein Kinase A (PKA) and Extracellular Signal-Regulated Kinase (ERK) have core roles in learning and memory. Here, we investigate kinase-kinase signaling interactions in the Drosophila brain Kenyon cell learning/memory circuit using separation of phases-based activity reporter of kinase (SPARK) biosensors to image circuit-localized functions in vivo. We find that constitutively active Rapidly Accelerated Fibrosarcoma (RAFgof) enhances ERK signaling only in Kenyon cell domains with low baseline PKA signaling, and that transgenic inhibition of PKA function elevates ERK signaling. Conversely, loss of ERK has no impact on PKA signaling, whereas RAFgof expands PKA signaling. Importantly, transgenic PKA inhibition together with RAFgof synergistically elevates ERK signaling. These findings indicate a negative PKA-ERK pathway interaction within learning/memory Kenyon cells. We find that potentiating circuit activity using an exogenous NaChBac ion channel elevates PKA signaling in circuit domains with low baseline PKA function, and uniformly strongly increases ERK signaling. Similarly, thermogenetic stimulation of circuit activity with a temperature-sensitive TRPA1 channel increases PKA signaling in circuit domains of low baseline PKA, and elevates ERK signaling. Importantly, potentiating circuit activity (NaChBac) while also inhibiting PKA function synergistically elevates ERK signaling. Likewise, conditional induction of circuit activity (TRPA1) together with PKA inhibition increases activity-dependent ERK signaling. Finally, a mechanically-induced seizure model (bang-sensitive sesB mutant) elevates PKA signaling, while simultaneous transgenic PKA inhibition in this model acts to synergistically increase ERK signaling. Taken together, we conclude PKA limits ERK signaling in Kenyon cells within the learning and memory circuit, with PKA function acting to restrict activity-dependent ERK signaling.
Collapse
Affiliation(s)
- James C Sears
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235, USA; Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA
| | - Kendal Broadie
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235, USA; Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235, USA; Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN 37235, USA; Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, TN 37235, USA; Vanderbilt Kennedy Center, Vanderbilt University and Medical Center, Nashville, TN 37235, USA.
| |
Collapse
|
2
|
Hwang HJ, Cox RT. Feeding a rich diet supplemented with the translation inhibitor cycloheximide decreases lifespan and ovary size in Drosophila. Biol Open 2024; 13:bio061697. [PMID: 39588711 PMCID: PMC11625892 DOI: 10.1242/bio.061697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/14/2024] [Indexed: 11/27/2024] Open
Abstract
Drosophila oogenesis has long been an important model for understanding myriad cellular processes controlling development, RNA biology and patterning. Flies are easily fed drugs to disrupt various molecular pathways. However, this is often done under poor nutrient conditions that adversely affect oogenesis, thus making analysis challenging. Cycloheximide is a widely used compound that binds to and stalls the ribosome, therefore reducing protein synthesis. As egg production is a highly nutrient-dependent process, we developed a method to feed female Drosophila a rich diet of yeast paste supplemented with cycloheximide to better determine the effect of cycloheximide treatment on oogenesis. We found that flies readily consumed cycloheximide-supplemented yeast paste. Males and females had reduced lifespans when maintained on cycloheximide, with males exhibiting a dose-dependent decrease. Although females did not exhibit decreased egg laying, their ovaries were smaller and the number of progeny reduced, indicating substandard egg quality. Finally, females fed cycloheximide had disrupted oogenesis, with smaller ovaries, missing ovariole stages, and an increase in apoptotic follicles. Together, these data support that reduced protein synthesis adversely affects oogenesis with a rich diet that provides optimal nutrient conditions. In addition, this method could be used more broadly to test the effect of other drugs on Drosophila oogenesis without the confounding effects caused by poor nutrition.
Collapse
Affiliation(s)
- Hye Jin Hwang
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, MD 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Rachel T. Cox
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, MD 20814, USA
| |
Collapse
|
3
|
Hwang HJ, Cox RT. Feeding a rich diet supplemented with the translation inhibitor cycloheximide decreases lifespan and ovary size in Drosophila. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608713. [PMID: 39411156 PMCID: PMC11475871 DOI: 10.1101/2024.08.19.608713] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Drosophila oogenesis has long been an important model for understanding myriad cellular processes controlling development, RNA biology, and patterning. Flies are easily fed drugs to disrupt various molecular pathways. However, this is often done under poor nutrient conditions that adversely affect oogenesis, thus making analysis challenging. Cycloheximide is a widely used compound that binds to and stalls the ribosome, therefore reducing protein synthesis. Since egg production is a highly nutrient-dependent process, we developed a method to feed female Drosophila a rich diet of yeast paste supplemented with cycloheximide to better determine the effect of cycloheximide treatment on oogenesis. We find flies readily consume cycloheximide-supplemented yeast paste. Males and females have reduced lifespans when maintained on cycloheximide, with males exhibiting a dose-dependent decrease. While females did not exhibit decreased egg-laying, their ovaries were smaller and the number of progeny reduced, indicating substandard egg quality. Finally, females fed cycloheximide have disrupted oogenesis, with smaller ovaries, missing ovariole stages, and an increase in apoptotic follicles. Together, these data support that reduced protein synthesis adversely affects oogenesis with a rich diet that provides optimal nutrient conditions. In addition, this method could be used more broadly to test the effect of other drugs on Drosophila oogenesis without the confounding effects caused by poor nutrition.
Collapse
Affiliation(s)
- Hye Jin Hwang
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, MD 20814
- Henry M. Jackson Foundation, Rockville, MD
| | - Rachel T. Cox
- Department of Biochemistry and Molecular Biology, Uniformed Services University, Bethesda, MD 20814
| |
Collapse
|
4
|
Sears JC, Broadie K. Use-Dependent, Untapped Dual Kinase Signaling Localized in Brain Learning Circuitry. J Neurosci 2024; 44:e1126232024. [PMID: 38267256 PMCID: PMC10957217 DOI: 10.1523/jneurosci.1126-23.2024] [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: 06/16/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/26/2024] Open
Abstract
Imaging brain learning and memory circuit kinase signaling is a monumental challenge. The separation of phases-based activity reporter of kinase (SPARK) biosensors allow circuit-localized studies of multiple interactive kinases in vivo, including protein kinase A (PKA) and extracellular signal-regulated kinase (ERK) signaling. In the precisely-mapped Drosophila brain learning/memory circuit, we find PKA and ERK signaling differentially enriched in distinct Kenyon cell connectivity nodes. We discover that potentiating normal circuit activity induces circuit-localized PKA and ERK signaling, expanding kinase function within new presynaptic and postsynaptic domains. Activity-induced PKA signaling shows extensive overlap with previously selective ERK signaling nodes, while activity-induced ERK signaling arises in new connectivity nodes. We find targeted synaptic transmission blockade in Kenyon cells elevates circuit-localized ERK induction in Kenyon cells with normally high baseline ERK signaling, suggesting lateral and feedback inhibition. We discover overexpression of the pathway-linking Meng-Po (human SBK1) serine/threonine kinase to improve learning acquisition and memory consolidation results in dramatically heightened PKA and ERK signaling in separable Kenyon cell circuit connectivity nodes, revealing both synchronized and untapped signaling potential. Finally, we find that a mechanically-induced epileptic seizure model (easily shocked "bang-sensitive" mutants) has strongly elevated, circuit-localized PKA and ERK signaling. Both sexes were used in all experiments, except for the hemizygous male-only seizure model. Hyperexcitable, learning-enhanced, and epileptic seizure models have comparably elevated interactive kinase signaling, suggesting a common basis of use-dependent induction. We conclude that PKA and ERK signaling modulation is locally coordinated in use-dependent spatial circuit dynamics underlying seizure susceptibility linked to learning/memory potential.
Collapse
Affiliation(s)
- James C Sears
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| | - Kendal Broadie
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Departments of Biological Sciences, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
- Vanderbilt Kennedy Center, Vanderbilt University and Medical Center, Nashville, Tennessee 37235
| |
Collapse
|
5
|
Loreto JS, Ferreira SA, de Almeida P, da Rocha JBT, Barbosa NV. Screening for Differentially Expressed Memory Genes on a Diabetes Model Induced by High-Sugar Diet in Drosophila melanogaster: Potential Markers for Memory Deficits. Mol Neurobiol 2024; 61:1225-1236. [PMID: 37698834 DOI: 10.1007/s12035-023-03598-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/18/2023] [Indexed: 09/13/2023]
Abstract
Type 2 diabetes mellitus (T2DM) has been shown to affect a series of cognitive processes including memory, increasing the risk for dementia, particularly Alzheimer's disease (AD). Although increasing evidence has supported that both diseases share common features, the pathophysiological mechanisms connecting these two disorders remain to be fully elucidated. Herein, we used Drosophila melanogaster fed on a high-sugar diet (HSD) to mimic T2DM, and investigate its effects on memory as well as identify potential molecular players associated with the memory deficits induced by HSD. Flies hatched from and reared on HSD for 7 days had a substantial decrease in short-term memory (STM). The screening for memory-related genes using transcriptome data revealed that HSD altered the expression of 33% of memory genes in relation to the control. Among the differentially expressed genes (DEGs) with a fold change (FC) higher than two, we found five genes, related to synapse and memory trace formation, that could be considered strong candidates to underlie the STM deficits in HSD flies: Abl tyrosine kinase (Abl), bruchpilot (Brp), minibrain (Mnb), shaker (Sh), and gilgamesh (Gish). We also analyzed genes from the dopamine system, one of the most relevant signaling pathways for olfactory memory. Interestingly, the flies fed on HSD presented a decreased expression of the Tyrosine hydroxylase (Ple) and Dopa decarboxylase (Ddc) genes, signals of a possible dopamine deficiency. In this work, we present promising biomarkers to investigate molecular networks shared between T2DM and AD.
Collapse
Affiliation(s)
- Julia Sepel Loreto
- Centro de Ciências Naturais E Exatas, Programa de Pós-Graduação Em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, Santa Maria, RS, 1000, 97105-900, Brazil
| | - Sabrina Antunes Ferreira
- Centro de Ciências Naturais E Exatas, Programa de Pós-Graduação Em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, Santa Maria, RS, 1000, 97105-900, Brazil
| | - Pâmela de Almeida
- Centro de Ciências Naturais E Exatas, Programa de Pós-Graduação Em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, Santa Maria, RS, 1000, 97105-900, Brazil
| | - João Batista Teixeira da Rocha
- Centro de Ciências Naturais E Exatas, Programa de Pós-Graduação Em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, Santa Maria, RS, 1000, 97105-900, Brazil
| | - Nilda Vargas Barbosa
- Centro de Ciências Naturais E Exatas, Programa de Pós-Graduação Em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Avenida Roraima, Santa Maria, RS, 1000, 97105-900, Brazil.
| |
Collapse
|
6
|
Hang M, Tse MCL, Pang BPS, Bi X, Jin F, Lee CW, Wong AOL, Chan CB. Differential regulation of hepatic SH3 domain binding kinase 1 (SBK1) expression in mouse and goldfish. Gen Comp Endocrinol 2023; 344:114372. [PMID: 37652166 DOI: 10.1016/j.ygcen.2023.114372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/26/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
SH3 domain binding kinase 1 (SBK1) is a serine/threonine kinase that belongs to the new kinase family (NFK) with limited information on its function. Previous studies reported that SBK1 plays a role in memory formation, lipid metabolism, and cancer cell progression. Nevertheless, the regulatory mechanism of Sbk1 expression in various tissues remains unknown. We report here that Sbk1 expression in mouse hepatocytes was downregulated by glucocorticoid, whereas saturated and unsaturated fatty acids were stimulators of Sbk1 expression. The regulatory role of glucocorticoid and fatty acid was further confirmed by the Sbk1 promoter assay, which aligned with the presence of several glucocorticoid-response elements (GRE) and peroxisome proliferator responsive elements (PPRE) in the mouse Sbk1 promoter. The inhibitory effect of glucocorticoids on hepatic Sbk1 expression and protein content could also be demonstrated in vivo after prednisolone injection. Moreover, the expression of SBK1 in goldfish (gfSBK1) was also sensitive to glucocorticoid suppression as their mouse orthologues. In contrast, insulin had a differential action on SBK1 expression that it promoted the expression of all SBK1 isoforms in the goldfish hepatocytes but inhibited Sbk1 expression in the mouse hepatocytes. Together, our findings indicate that SBK1 expression is hormone- and nutrient-sensitive with a species-specific response.
Collapse
Affiliation(s)
- Miaojia Hang
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Margaret Chui Ling Tse
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Brian Pak Shing Pang
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Xinyi Bi
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Fanming Jin
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Wai Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Anderson O L Wong
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Chi Bun Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region.
| |
Collapse
|
7
|
Frecot DI, Froehlich T, Rothbauer U. 30 years of nanobodies - an ongoing success story of small binders in biological research. J Cell Sci 2023; 136:jcs261395. [PMID: 37937477 DOI: 10.1242/jcs.261395] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023] Open
Abstract
A milestone in the field of recombinant binding molecules was achieved 30 years ago with the discovery of single-domain antibodies from which antigen-binding variable domains, better known as nanobodies (Nbs), can be derived. Being only one tenth the size of conventional antibodies, Nbs feature high affinity and specificity, while being highly stable and soluble. In addition, they display accessibility to cryptic sites, low off-target accumulation and deep tissue penetration. Efficient selection methods, such as (semi-)synthetic/naïve or immunized cDNA libraries and display technologies, have facilitated the isolation of Nbs against diverse targets, and their single-gene format enables easy functionalization and high-yield production. This Review highlights recent advances in Nb applications in various areas of biological research, including structural biology, proteomics and high-resolution and in vivo imaging. In addition, we provide insights into intracellular applications of Nbs, such as live-cell imaging, biosensors and targeted protein degradation.
Collapse
Affiliation(s)
- Desiree I Frecot
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Markwiesenstrasse 55, 72770 Reutlingen, Reutlingen, Germany
| | - Theresa Froehlich
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| | - Ulrich Rothbauer
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, 72076 Tuebingen, Germany
| |
Collapse
|
8
|
Chen CC, Lin HW, Feng KL, Tseng DW, de Belle JS, Chiang AS. A subset of cholinergic mushroom body neurons blocks long-term memory formation in Drosophila. Cell Rep 2023; 42:112974. [PMID: 37590142 DOI: 10.1016/j.celrep.2023.112974] [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: 07/23/2022] [Revised: 12/22/2022] [Accepted: 07/29/2023] [Indexed: 08/19/2023] Open
Abstract
Long-term memory (LTM) requires learning-induced synthesis of new proteins allocated to specific neurons and synapses in a neural circuit. Not all learned information, however, becomes permanent memory. How the brain gates relevant information into LTM remains unclear. In Drosophila adults, weak learning after a single training session in an olfactory aversive task typically does not induce protein-synthesis-dependent LTM. Instead, strong learning after multiple spaced training sessions is required. Here, we report that pre-synaptic active-zone protein synthesis and cholinergic signaling from the early α/β subset of mushroom body (MB) neurons produce a downstream inhibitory effect on LTM formation. When we eliminated inhibitory signaling from these neurons, weak learning was then sufficient to form LTM. This bidirectional circuit mechanism modulates the transition between distinct memory phase functions in different subpopulations of MB neurons in the olfactory memory circuit.
Collapse
Affiliation(s)
- Chun-Chao Chen
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.
| | - Hsuan-Wen Lin
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kuan-Lin Feng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Der-Wan Tseng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - J Steven de Belle
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA; Department of Psychological Sciences, University of San Diego, San Diego, CA 92110, USA; School of Life Sciences, University of Nevada, Las Vegas, NV 89154, USA; MnemOdyssey LLC, Escondido, CA 92027, USA
| | - Ann-Shyn Chiang
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan; Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 80780, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Clinical Medical Science, China Medical University, Taichung 40402, Taiwan; Kavli Institute for Brain and Mind, University of California at San Diego, La Jolla, CA 92093-0526, USA.
| |
Collapse
|
9
|
Davis RL. Learning and memory using Drosophila melanogaster: a focus on advances made in the fifth decade of research. Genetics 2023; 224:iyad085. [PMID: 37212449 PMCID: PMC10411608 DOI: 10.1093/genetics/iyad085] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023] Open
Abstract
In the last decade, researchers using Drosophila melanogaster have made extraordinary progress in uncovering the mysteries underlying learning and memory. This progress has been propelled by the amazing toolkit available that affords combined behavioral, molecular, electrophysiological, and systems neuroscience approaches. The arduous reconstruction of electron microscopic images resulted in a first-generation connectome of the adult and larval brain, revealing complex structural interconnections between memory-related neurons. This serves as substrate for future investigations on these connections and for building complete circuits from sensory cue detection to changes in motor behavior. Mushroom body output neurons (MBOn) were discovered, which individually forward information from discrete and non-overlapping compartments of the axons of mushroom body neurons (MBn). These neurons mirror the previously discovered tiling of mushroom body axons by inputs from dopamine neurons and have led to a model that ascribes the valence of the learning event, either appetitive or aversive, to the activity of different populations of dopamine neurons and the balance of MBOn activity in promoting avoidance or approach behavior. Studies of the calyx, which houses the MBn dendrites, have revealed a beautiful microglomeruluar organization and structural changes of synapses that occur with long-term memory (LTM) formation. Larval learning has advanced, positioning it to possibly lead in producing new conceptual insights due to its markedly simpler structure over the adult brain. Advances were made in how cAMP response element-binding protein interacts with protein kinases and other transcription factors to promote the formation of LTM. New insights were made on Orb2, a prion-like protein that forms oligomers to enhance synaptic protein synthesis required for LTM formation. Finally, Drosophila research has pioneered our understanding of the mechanisms that mediate permanent and transient active forgetting, an important function of the brain along with acquisition, consolidation, and retrieval. This was catalyzed partly by the identification of memory suppressor genes-genes whose normal function is to limit memory formation.
Collapse
Affiliation(s)
- Ronald L Davis
- Department of Neuroscience, Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, University of Florida, 130 Scripps Way, Jupiter, FL 33458, USA
| |
Collapse
|
10
|
Chen X, Sun Z, Zhou S, Jiang W, Li J, Song G, Zhu X. SH3 domain-binding kinase 1 promotes proliferation and inhibits apoptosis of cervical cancer via activating the Wnt/β-catenin and Raf/ERK1/2 signaling pathways. Mol Carcinog 2023; 62:1147-1162. [PMID: 37132991 DOI: 10.1002/mc.23552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/27/2023] [Accepted: 04/20/2023] [Indexed: 05/04/2023]
Abstract
SH3 domain-binding kinase 1 (SBK1), is a member of the serine/threonine protein kinases family, and was confirmed to be upregulated in cervical cancer in our previous study. Nonetheless, the role of SBK1 in regulating cancer occurrence and development is unclear. In this study, the stable SBK1-knockdown and -overexpressed cell models were constructed by plasmid transfection technology. Cell viability and growth were assessed through CCK-8, colony formation, and BrdU methods. Cell cycle and apoptosis were analyzed by flow cytometry. The JC-1 staining assay was used to explore mitochondrial membrane potential. The scratch and Transwell assays were used to evaluate the cell metastatic ability. The nude mice models were utilized to explore the SBK1 expression affecting tumor growth in vivo. Our research indicated a high expression of SBK1 both in tissues and cells of cervical cancer. The proliferative, migratory, as well as invasive capacities of cervical cancer cells, were suppressed, and apoptosis was enhanced after SBK1 silence, whereas SBK1 upregulation led to opposite results. In addition, Wnt/β-catenin and Raf/ERK1/2 pathways were activated by SBK1 upregulation. Furthermore, downregulation of c-Raf or β-catenin, reversed the proliferation promotion and apoptosis inhibition effects in SBK1-overexpressed cells. The same results were observed with the use of the specific Raf inhibitor. SBK1 overexpression also contributed to tumor growth in vivo. Overall, SBK1 played a vital role in cervical tumorigenesis via activating the Wnt/β-catenin and Raf/ERK1/2 pathways.
Collapse
Affiliation(s)
- Xin Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhengwei Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shengjie Zhou
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Wenxiao Jiang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jieyi Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gendi Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xueqiong Zhu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Obstetrics and Gynecology, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou, Zhejiang, China
| |
Collapse
|
11
|
Rozich E, Randolph LK, Insolera R. An optimized temporally controlled Gal4 system in Drosophila reveals degeneration caused by adult-onset neuronal Vps13D knockdown. Front Neurosci 2023; 17:1204068. [PMID: 37457002 PMCID: PMC10339317 DOI: 10.3389/fnins.2023.1204068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/09/2023] [Indexed: 07/18/2023] Open
Abstract
Mutations in the human gene VPS13D cause the adult-onset neurodegenerative disease ataxia. Our previous work showed that disruptions in the Vps13D gene in Drosophila neurons causes mitochondrial defects. However, developmental lethality caused by Vps13D loss limited our understanding of the long-term physiological effects of Vps13D perturbation in neurons. Here, we optimized a previously generated system to temporally knock down Vps13D expression precisely in adult Drosophila neurons using a modification to the Gal4/UAS system. Adult-onset activation of Gal4 was enacted using the chemically-inducible tool which fuses a destabilization-domain to the Gal4 repressor Gal80 (Gal80-DD). Optimization of the Gal80-DD tool shows that feeding animals the DD-stabilizing drug trimethoprim (TMP) during development and rearing at a reduced temperature maximally represses Gal4 activity. Temperature shift and removal of TMP from the food after eclosion robustly activates Gal4 expression in adult neurons. Using the optimized Gal80-DD system, we find that adult-onset Vps13D RNAi expression in neurons causes the accumulation of mitophagy intermediates, progressive deficits in locomotor activity, early lethality, and brain vacuolization characteristic of neurodegeneration. The development of this optimized system allows us to more precisely examine the degenerative phenotypes caused by Vps13D disruption, and can likely be utilized in the future for other genes associated with neurological diseases whose manipulation causes developmental lethality in Drosophila.
Collapse
Affiliation(s)
- Emily Rozich
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| | - Lynsey K. Randolph
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, United States
| | - Ryan Insolera
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI, United States
| |
Collapse
|
12
|
Ahuja P, Bi X, Ng CF, Tse MCL, Hang M, Pang BPS, Iu ECY, Chan WS, Ooi XC, Sun A, Herlea-Pana O, Liu Z, Yang X, Jiao B, Ma X, Wu KKL, Lee LTO, Cheng KKY, Lee CW, Chan CB. Src homology 3 domain binding kinase 1 protects against hepatic steatosis and insulin resistance through the Nur77-FGF21 pathway. Hepatology 2023; 77:213-229. [PMID: 35363898 DOI: 10.1002/hep.32501] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 02/03/2023]
Abstract
BACKGROUND AND AIMS Metabolism in the liver is dysregulated in obesity, contributing to various health problems including steatosis and insulin resistance. While the pathogenesis of lipid accumulation has been extensively studied, the protective mechanism against lipid challenge in the liver remains unclear. Here, we report that Src homology 3 domain binding kinase 1 (SBK1) is a regulator of hepatic lipid metabolism and systemic insulin sensitivity in response to obesity. APPROACH AND RESULTS Enhanced Sbk1 expression was found in the liver of high-fat diet (HFD)-induced obese mice and fatty acid (FA)-challenged hepatocytes. SBK1 knockdown in mouse liver cells augmented FA uptake and lipid accumulation. Similarly, liver-specific SBK1 knockout ( Lsko ) mice displayed more severe hepatosteatosis and higher expression of genes in FA uptake and lipogenesis than the Flox/Flox ( Fl/Fl ) control mice when fed the HFD. The HFD-fed Lsko mice also showed symptoms of hyperglycemia, poor systemic glucose tolerance, and lower insulin sensitivity than the Fl/Fl mice. On the other hand, hepatic Sbk1 overexpression alleviated the high-fructose diet-induced hepatosteatosis, hyperlipidemia, and hyperglycemia in mice. White adipose tissue browning was also observed in hepatic SBK1 -overexpressed mice. Moreover, we found that SBK1 was a positive regulator of FGF21 in the liver during energy surplus conditions. Mechanistically, SBK1 phosphorylates the orphan nuclear receptor 4A1 (Nur77) on serine 344 to promote hepatic FGF21 expression and inhibit the transcription of genes involved in lipid anabolism. CONCLUSIONS Collectively, our data suggest that SBK1 is a regulator of the metabolic adaption against obesity through the Nur77-FGF21 pathway.
Collapse
Affiliation(s)
- Palak Ahuja
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Xinyi Bi
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Chun Fai Ng
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | | | - Miaojia Hang
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Brian Pak Shing Pang
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Elsie Chit Yu Iu
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Wing Suen Chan
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Xin Ci Ooi
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Anqi Sun
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Oana Herlea-Pana
- Department of Physiology , The University of Oklahoma Health Sciences Center , Oklahoma City , Oklahoma , USA
| | - Zhixue Liu
- Center for Molecular & Translational Medicine , Georgia State University , Atlanta , Georgia , USA
| | - Xiuying Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines and Beijing , Key Laboratory of Drug Target and Screening Research , Institute of Materia Medica of Peking Union Medical College , Beijing , China
| | - Baowei Jiao
- State Key Laboratory of Genetic Resources and Evolution , Kunming Institute of Zoology , Chinese Academy of Sciences , Kunming , China
| | - Xin Ma
- Cancer Centre , Faculty of Health Sciences , University of Macau , Taipa, Macau , China
| | - Kelvin Ka Lok Wu
- Department of Health Technology and Informatics , The Hong Kong Polytechnic University , Hong Kong SAR , China
| | - Leo Tsz On Lee
- Cancer Centre , Faculty of Health Sciences , University of Macau , Taipa, Macau , China
- MOE Frontiers Science Center for Precision Oncology , University of Macau , Taipa, Macau , China
| | - Kenneth King Yip Cheng
- Department of Health Technology and Informatics , The Hong Kong Polytechnic University , Hong Kong SAR , China
| | - Chi Wai Lee
- School of Biomedical Sciences , The University of Hong Kong , Hong Kong SAR , China
| | - Chi Bun Chan
- School of Biological Sciences , The University of Hong Kong , Hong Kong SAR , China
- State Key Laboratory of Pharmaceutical Biotechnology , The University of Hong Kong , Hong Kong SAR , China
| |
Collapse
|
13
|
CREBB repression of protein synthesis in mushroom body gates long-term memory formation in Drosophila. Proc Natl Acad Sci U S A 2022; 119:e2211308119. [PMID: 36469774 PMCID: PMC9897441 DOI: 10.1073/pnas.2211308119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Learned experiences are not necessarily consolidated into long-term memory (LTM) unless they are periodic and meaningful. LTM depends on de novo protein synthesis mediated by cyclic AMP response element-binding protein (CREB) activity. In Drosophila, two creb genes (crebA, crebB) and multiple CREB isoforms have reported influences on aversive olfactory LTM in response to multiple cycles of spaced conditioning. How CREB isoforms regulate LTM effector genes in various neural elements of the memory circuit is unclear, especially in the mushroom body (MB), a prominent associative center in the fly brain that has been shown to participate in LTM formation. Here, we report that i) spaced training induces crebB expression in MB α-lobe neurons and ii) elevating specific CREBB isoform levels in the early α/β subpopulation of MB neurons enhances LTM formation. By contrast, learning from weak training iii) induces 5-HT1A serotonin receptor synthesis, iv) activates 5-HT1A in early α/β neurons, and v) inhibits LTM formation. vi) LTM is enhanced when this inhibitory effect is relieved by down-regulating 5-HT1A or overexpressing CREBB. Our findings show that spaced training-induced CREBB antagonizes learning-induced 5-HT1A in early α/β MB neurons to modulate LTM consolidation.
Collapse
|
14
|
Chen J, Zhu H, Wang R, Su X, Ruan Z, Pan Y, Peng Q. Functional Dissection of Protein Kinases in Sexual Development and Female Receptivity of Drosophila. Front Cell Dev Biol 2022; 10:923171. [PMID: 35757001 PMCID: PMC9220291 DOI: 10.3389/fcell.2022.923171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Protein phosphorylation is crucial for a variety of biological functions, but how it is involved in sexual development and behavior is rarely known. In this study, we performed a screen of RNA interference targeting 177 protein kinases in Drosophila and identified 13 kinases involved in sexual development in one or both sexes. We further identified that PKA and CASK promote female sexual behavior while not affecting female differentiation. Knocking down PKA or CASK in about five pairs of pC1 neurons in the central brain affects the fine projection but not cell number of these pC1 neurons and reduces virgin female receptivity. We also found that PKA and CASK signaling is required acutely during adulthood to promote female sexual behavior. These results reveal candidate kinases required for sexual development and behaviors and provide insights into how kinases would regulate neuronal development and physiology to fine tune the robustness of sexual behaviors.
Collapse
Affiliation(s)
- Jiangtao Chen
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Huan Zhu
- School of Biological and Medical Engineering, Southeast University, Nanjing, China
| | - Rong Wang
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Xiangbin Su
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Zongcai Ruan
- School of Biological and Medical Engineering, Southeast University, Nanjing, China
| | - Yufeng Pan
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| | - Qionglin Peng
- The Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing, China
| |
Collapse
|
15
|
Andreani T, Rosensweig C, Sisobhan S, Ogunlana E, Kath W, Allada R. Circadian programming of the ellipsoid body sleep homeostat in Drosophila. eLife 2022; 11:e74327. [PMID: 35735904 PMCID: PMC9270026 DOI: 10.7554/elife.74327] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 06/23/2022] [Indexed: 11/18/2022] Open
Abstract
Homeostatic and circadian processes collaborate to appropriately time and consolidate sleep and wake. To understand how these processes are integrated, we scheduled brief sleep deprivation at different times of day in Drosophila and find elevated morning rebound compared to evening. These effects depend on discrete morning and evening clock neurons, independent of their roles in circadian locomotor activity. In the R5 ellipsoid body sleep homeostat, we identified elevated morning expression of activity dependent and presynaptic gene expression as well as the presynaptic protein BRUCHPILOT consistent with regulation by clock circuits. These neurons also display elevated calcium levels in response to sleep loss in the morning, but not the evening consistent with the observed time-dependent sleep rebound. These studies reveal the circuit and molecular mechanisms by which discrete circadian clock neurons program a homeostatic sleep center.
Collapse
Affiliation(s)
- Tomas Andreani
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Clark Rosensweig
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Shiju Sisobhan
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - Emmanuel Ogunlana
- Department of Neurobiology, Northwestern UniversityEvanstonUnited States
| | - William Kath
- Department of Engineering Sciences and Applied Mathematics, Northwestern UniversityEvanstonUnited States
| | - Ravi Allada
- Department of Neurobiology, Northwestern UniversityChicagoUnited States
| |
Collapse
|
16
|
van Gorp PRR, Zhang J, Liu J, Tsonaka R, Mei H, Dekker SO, Bart CI, De Coster T, Post H, Heck AJR, Schalij MJ, Atsma DE, Pijnappels DA, de Vries AAF. Sbk2, a Newly Discovered Atrium-Enriched Regulator of Sarcomere Integrity. Circ Res 2022; 131:24-41. [PMID: 35587025 DOI: 10.1161/circresaha.121.319300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Heart development relies on tight spatiotemporal control of cardiac gene expression. Genes involved in this intricate process have been identified using animals and pluripotent stem cell-based models of cardio(myo)genesis. Recently, the repertoire of cardiomyocyte differentiation models has been expanded with iAM-1, a monoclonal line of conditionally immortalized neonatal rat atrial myocytes (NRAMs), which allows toggling between proliferative and differentiated (ie, excitable and contractile) phenotypes in a synchronized and homogenous manner. METHODS In this study, the unique properties of conditionally immortalized NRAMs (iAMs) were exploited to identify and characterize (lowly expressed) genes with an as-of-yet uncharacterized role in cardiomyocyte differentiation. RESULTS Transcriptome analysis of iAM-1 cells at different stages during one cycle of differentiation and subsequent dedifferentiation identified ≈13 000 transcripts, of which the dynamic changes in expression upon cardiomyogenic differentiation mostly opposed those during dedifferentiation. Among the genes whose expression increased during differentiation and decreased during dedifferentiation were many with known (lineage-specific) functions in cardiac muscle formation. Filtering for cardiac-enriched low-abundance transcripts, identified multiple genes with an uncharacterized role during cardio(myo)genesis including Sbk2 (SH3 domain binding kinase family member 2). Sbk2 encodes an evolutionarily conserved putative serine/threonine protein kinase, whose expression is strongly up- and downregulated during iAM-1 cell differentiation and dedifferentiation, respectively. In neonatal and adult rats, the protein is muscle-specific, highly atrium-enriched, and localized around the A-band of cardiac sarcomeres. Knockdown of Sbk2 expression caused loss of sarcomeric organization in NRAMs, iAMs and their human counterparts, consistent with a decrease in sarcomeric gene expression as evinced by transcriptome and proteome analyses. Interestingly, co-immunoprecipitation using Sbk2 as bait identified possible interaction partners with diverse cellular functions (translation, intracellular trafficking, cytoskeletal organization, chromatin modification, sarcomere formation). CONCLUSIONS iAM-1 cells are a relevant and suitable model to identify (lowly expressed) genes with a hitherto unidentified role in cardiomyocyte differentiation as exemplified by Sbk2: a regulator of atrial sarcomerogenesis.
Collapse
Affiliation(s)
- P R R van Gorp
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - J Zhang
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - J Liu
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.).,Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, the Netherlands. (H.M.)
| | - R Tsonaka
- Department of Biomedical Data Sciences, Medical Statistics Section, Leiden University Medical Center, the Netherlands. (R.T.)
| | - H Mei
- Central Laboratory, Longgang District People's Hospital of Shenzhen & The Third Affiliated Hospital of The Chinese University of Hong Kong, China (J.L.)
| | - S O Dekker
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - C I Bart
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - T De Coster
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - H Post
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, the Netherlands (H.P., A.J.R.H.).,Netherlands Proteomics Centre, the Netherlands (H.P., A.J.R.H.)
| | - A J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, the Netherlands (H.P., A.J.R.H.).,Netherlands Proteomics Centre, the Netherlands (H.P., A.J.R.H.)
| | - M J Schalij
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - D E Atsma
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - D A Pijnappels
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| | - A A F de Vries
- Laboratory of Experimental Cardiology, Department of Cardiology, Leiden University Medical Center, the Netherlands. (P.R.R.v.G., J.Z., J.L., S.O.D., C.I.B., T.D.C., M.J.S., D.E.A., D.A.P., A.A.F.d.V.)
| |
Collapse
|
17
|
MicroRNA-377-3p promotes cell proliferation and inhibits cell cycle arrest and cell apoptosis in hepatocellular carcinoma by affecting EGR1-mediated p53 activation. Pathol Res Pract 2022; 234:153855. [PMID: 35461040 DOI: 10.1016/j.prp.2022.153855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 03/14/2022] [Accepted: 03/22/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an aggressive malignant carcinoma with a high fatality rate. MicroRNAs (miRNAs) have been found to regulate the development of multiple cancers, including HCC. MATERIALS AND METHODS Quantitative polymerase chain reaction (qPCR) were implemented to evaluate RNA level and western blot to detect protein level. Cell counting kit-8 (CCK-8), terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL), flow cytometry and in vivo assays were performed to evaluate the biological functions of RNAs on HCC cell proliferation, cell cycle and apoptosis. Luciferase reporter gene and chromatin immunoprecipitation (ChIP) assays were carried out to evaluate the underlying mechanisms. RESULTS MiR-377-3p promotes cell proliferation and inhibits cell cycle arrest and cell apoptosis in HCC. MiR-377-3p downregulates transcription factor EGR1 expression to weaken the activation of p53. p53 inhibits CCNB1, CCNB2 and CHEK1 expressions and activates THBS1, IGFBP3 and TRIM22 expressions. p53 knockdown promotes the proliferation and inhibits the cell cycle arrest and apoptosis of HCC cells. CONCLUSION Our study demonstrated the role and underlying mechanisms of miR-377-3p in HCC. MiR-377-3p facilitates the proliferation and suppresses the cell cycle arrest and apoptosis in HCC by affecting transcription factor EGR1-mediated p53 activation.
Collapse
|
18
|
Sears JC, Broadie K. Temporally and Spatially Localized PKA Activity within Learning and Memory Circuitry Regulated by Network Feedback. eNeuro 2022; 9:ENEURO.0450-21.2022. [PMID: 35301221 PMCID: PMC8982635 DOI: 10.1523/eneuro.0450-21.2022] [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: 10/24/2021] [Revised: 02/18/2022] [Accepted: 03/12/2022] [Indexed: 12/02/2022] Open
Abstract
Dynamic functional connectivity within brain circuits requires coordination of intercellular signaling and intracellular signal transduction. Critical roles for cAMP-dependent protein kinase A (PKA) signaling are well established in the Drosophila mushroom body (MB) learning and memory circuitry, but local PKA activity within this well-mapped neuronal network is uncharacterized. Here, we use an in vivo PKA activity sensor (PKA-SPARK) to test spatiotemporal regulatory requirements in the MB axon lobes. We find immature animals have little detectable PKA activity, whereas postcritical period adults show high field-selective activation primarily in just 3/16 defined output regions. In addition to the age-dependent PKA activity in distinct α'/β' lobe nodes, females show sex-dependent elevation compared with males in these same restricted regions. Loss of neural cell body Fragile X mental retardation protein (FMRP) and Rugose [human Neurobeachin (NBEA)] suppresses localized PKA activity, whereas overexpression (OE) of MB lobe PKA-synergist Meng-Po (human SBK1) promotes PKA activity. Elevated Meng-Po subverts the PKA age-dependence, with elevated activity in immature animals, and spatial-restriction, with striking γ lobe activity. Testing circuit signaling requirements with temperature-sensitive shibire (human Dynamin) blockade, we find broadly expanded PKA activity within the MB lobes. Using transgenic tetanus toxin to block MB synaptic output, we find greatly heightened PKA activity in virtually all MB lobe fields, although the age-dependence is maintained. We conclude spatiotemporally restricted PKA activity signaling within this well-mapped learning/memory circuit is age-dependent and sex-dependent, driven by FMRP-Rugose pathway activation, temporally promoted by Meng-Po kinase function, and restricted by output neurotransmission providing network feedback.
Collapse
Affiliation(s)
- James C Sears
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235
| | - Kendal Broadie
- Department of Biological Sciences, Vanderbilt University and Medical Center, Nashville, TN 37235
- Vanderbilt Brain Institute, Vanderbilt University and Medical Center, Nashville, TN 37235
- Department of Cell and Developmental Biology, Vanderbilt University and Medical Center, Nashville, TN 37235
- Department of Pharmacology, Vanderbilt University and Medical Center, Nashville, TN 37235
| |
Collapse
|
19
|
Marcogliese PC, Dutta D, Ray SS, Dang NDP, Zuo Z, Wang Y, Lu D, Fazal F, Ravenscroft TA, Chung H, Kanca O, Wan J, Douine ED, Network UD, Pena LDM, Yamamoto S, Nelson SF, Might M, Meyer KC, Yeo NC, Bellen HJ. Loss of IRF2BPL impairs neuronal maintenance through excess Wnt signaling. SCIENCE ADVANCES 2022; 8:eabl5613. [PMID: 35044823 PMCID: PMC8769555 DOI: 10.1126/sciadv.abl5613] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/30/2021] [Indexed: 05/12/2023]
Abstract
De novo truncations in Interferon Regulatory Factor 2 Binding Protein Like (IRF2BPL) lead to severe childhood-onset neurodegenerative disorders. To determine how loss of IRF2BPL causes neural dysfunction, we examined its function in Drosophila and zebrafish. Overexpression of either IRF2BPL or Pits, the Drosophila ortholog, represses Wnt transcription in flies. In contrast, neuronal depletion of Pits leads to increased wingless (wg) levels in the brain and is associated with axonal loss, whereas inhibition of Wg signaling is neuroprotective. Moreover, increased neuronal expression of wg in flies is sufficient to cause age-dependent axonal loss, similar to reduction of Pits. Loss of irf2bpl in zebrafish also causes neurological defects with an associated increase in wnt1 transcription and downstream signaling. WNT1 is also increased in patient-derived astrocytes, and pharmacological inhibition of Wnt suppresses the neurological phenotypes. Last, IRF2BPL and the Wnt antagonist, CKIα, physically and genetically interact, showing that IRF2BPL and CkIα antagonize Wnt transcription and signaling.
Collapse
Affiliation(s)
- Paul C. Marcogliese
- 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
| | - Debdeep Dutta
- 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
| | - Shrestha Sinha Ray
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - Nghi D. P. Dang
- Department of Pharmacology and Toxicology, University of Alabama, Birmingham, AL 35294, USA
| | - Zhongyuan Zuo
- 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
| | - Yuchun Wang
- 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
| | - Di Lu
- 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
| | - Fatima Fazal
- 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
| | - Thomas A. Ravenscroft
- 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
| | - Hyunglok Chung
- 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
| | - Oguz Kanca
- 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
| | - JiJun Wan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Emilie D. Douine
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Undiagnosed Diseases Network
- 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
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pharmacology and Toxicology, University of Alabama, Birmingham, AL 35294, USA
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, USA
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
- Precision Medicine Institute, University of Alabama, Birmingham, AL 35294, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Loren D. M. Pena
- Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45267, 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 Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Development, Disease Models & Therapeutics Graduate Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stanley F. Nelson
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Matthew Might
- Precision Medicine Institute, University of Alabama, Birmingham, AL 35294, USA
| | - Kathrin C. Meyer
- The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, OH 43210, USA
| | - Nan Cher Yeo
- Department of Pharmacology and Toxicology, University of Alabama, Birmingham, AL 35294, USA
- Precision Medicine Institute, University of Alabama, Birmingham, AL 35294, 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
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
20
|
Feng KL, Weng JY, Chen CC, Abubaker MB, Lin HW, Charng CC, Lo CC, de Belle JS, Tully T, Lien CC, Chiang AS. Neuropeptide F inhibits dopamine neuron interference of long-term memory consolidation in Drosophila. iScience 2021; 24:103506. [PMID: 34934925 DOI: 10.1016/j.isci.2021.103506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/13/2021] [Accepted: 11/22/2021] [Indexed: 11/28/2022] Open
Abstract
Long-term memory (LTM) formation requires consolidation processes to overcome interfering signals that erode memory formation. Olfactory memory in Drosophila involves convergent projection neuron (PN; odor) and dopaminergic neuron (DAN; reinforcement) input to the mushroom body (MB). How post-training DAN activity in the posterior lateral protocerebrum (PPL1) continues to regulate memory consolidation remains unknown. Here we address this question using targeted transgenes in behavior and electrophysiology experiments to show that (1) persistent post-training activity of PPL1-α2α'2 and PPL1-α3 DANs interferes with aversive LTM formation; (2) neuropeptide F (NPF) signaling blocks this interference in PPL1-α2α'2 and PPL1-α3 DANs after spaced training to enable LTM formation; and (3) training-induced NPF release and neurotransmission from two upstream dorsal-anterior-lateral (DAL2) neurons are required to form LTM. Thus, NPF signals from DAL2 neurons to specific PPL1 DANs disinhibit the memory circuit, ensuring that periodic events are remembered as consolidated LTM.
Collapse
Affiliation(s)
- Kuan-Lin Feng
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ju-Yun Weng
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chun-Chao Chen
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | | | - Hsuan-Wen Lin
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ching-Che Charng
- Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chung-Chuan Lo
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.,Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - J Steven de Belle
- Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.,Department of Psychological Sciences, University of San Diego, San Diego, CA 92110, USA.,School of Life Sciences, University of Nevada, Las Vegas, NV 89154, USA.,MnemOdyssey LLC, Escondido, CA 92027, USA
| | - Tim Tully
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Cheng-Chang Lien
- Institute of Neuroscience and Brain Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Ann-Shyn Chiang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu 30013, Taiwan.,Brain Research Center, National Tsing Hua University, Hsinchu 30013, Taiwan.,Institute of Systems Neuroscience and Department of Life Science, National Tsing Hua University, Hsinchu 30013, Taiwan.,Kaohsiung Medical University, Kaohsiung 80708, Taiwan.,National Health Research Institutes, Zhunan 35053, Taiwan.,China Medical University, Taichung 40402, Taiwan.,Kavli Institute for Brain and Mind, University of California at San Diego, La Jolla, CA 92093-0526, USA
| |
Collapse
|
21
|
Jiang Y, Hao N. Memorizing environmental signals through feedback and feedforward loops. Curr Opin Cell Biol 2021; 69:96-102. [PMID: 33549848 PMCID: PMC8058236 DOI: 10.1016/j.ceb.2020.11.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/25/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022]
Abstract
Cells in diverse organisms can store the information of previous environmental conditions for long periods of time. This form of cellular memory adjusts the cell's responses to future challenges, providing fitness advantages in fluctuating environments. Many biological functions, including cellular memory, are mediated by specific recurring patterns of interactions among proteins and genes, known as 'network motifs.' In this review, we focus on three well-characterized network motifs - negative feedback loops, positive feedback loops, and feedforward loops, which underlie different types of cellular memories. We describe the latest studies identifying these motifs in various molecular processes and discuss how the topologies and dynamics of these motifs can enable memory encoding and storage.
Collapse
Affiliation(s)
- Yanfei Jiang
- Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Nan Hao
- Section of Molecular Biology, Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
22
|
Kanca O, Zirin J, Garcia-Marques J, Knight SM, Yang-Zhou D, Amador G, Chung H, Zuo Z, Ma L, He Y, Lin WW, Fang Y, Ge M, Yamamoto S, Schulze KL, Hu Y, Spradling AC, Mohr SE, Perrimon N, Bellen HJ. An efficient CRISPR-based strategy to insert small and large fragments of DNA using short homology arms. eLife 2019; 8:e51539. [PMID: 31674908 PMCID: PMC6855806 DOI: 10.7554/elife.51539] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/31/2019] [Indexed: 12/31/2022] Open
Abstract
We previously reported a CRISPR-mediated knock-in strategy into introns of Drosophila genes, generating an attP-FRT-SA-T2A-GAL4-polyA-3XP3-EGFP-FRT-attP transgenic library for multiple uses (Lee et al., 2018a). The method relied on double stranded DNA (dsDNA) homology donors with ~1 kb homology arms. Here, we describe three new simpler ways to edit genes in flies. We create single stranded DNA (ssDNA) donors using PCR and add 100 nt of homology on each side of an integration cassette, followed by enzymatic removal of one strand. Using this method, we generated GFP-tagged proteins that mark organelles in S2 cells. We then describe two dsDNA methods using cheap synthesized donors flanked by 100 nt homology arms and gRNA target sites cloned into a plasmid. Upon injection, donor DNA (1 to 5 kb) is released from the plasmid by Cas9. The cassette integrates efficiently and precisely in vivo. The approach is fast, cheap, and scalable.
Collapse
Affiliation(s)
- Oguz Kanca
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Jonathan Zirin
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | | | - Shannon Marie Knight
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Donghui Yang-Zhou
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Gabriel Amador
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Hyunglok Chung
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Zhongyuan Zuo
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
| | - Liwen Ma
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Yuchun He
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Howard Hughes Medical Institute, Baylor College of MedicineHoustonUnited States
| | - Wen-Wen Lin
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Ying Fang
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Ming Ge
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Shinya Yamamoto
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Program in Developmental BiologyBaylor College of MedicineHoustonUnited States
- Department of NeuroscienceBaylor College of MedicineHoustonUnited States
| | - Karen L Schulze
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Howard Hughes Medical Institute, Baylor College of MedicineHoustonUnited States
| | - Yanhui Hu
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Allan C Spradling
- Department of EmbryologyHoward Hughes Medical Institute, Carnegie Institution for ScienceBaltimoreUnited States
| | - Stephanie E Mohr
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Norbert Perrimon
- Howard Hughes Medical Institute, Harvard Medical SchoolBostonUnited States
- Department of GeneticsHarvard Medical SchoolBostonUnited States
| | - Hugo J Bellen
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research Institute, Texas Children’s HospitalHoustonUnited States
- Howard Hughes Medical Institute, Baylor College of MedicineHoustonUnited States
- Program in Developmental BiologyBaylor College of MedicineHoustonUnited States
- Department of NeuroscienceBaylor College of MedicineHoustonUnited States
| |
Collapse
|
23
|
Alleman A, Stoldt M, Feldmeyer B, Foitzik S. Tandem-running and scouting behaviour are characterized by up-regulation of learning and memory formation genes within the ant brain. Mol Ecol 2019; 28:2342-2359. [PMID: 30903719 DOI: 10.1111/mec.15079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 03/09/2019] [Accepted: 03/12/2019] [Indexed: 12/13/2022]
Abstract
Tandem-running is a recruitment behaviour in ants that has been described as a form of teaching, where spatial information possessed by a leader is conveyed to following nestmates. Within Temnothorax ants, tandem-running is used within a variety of contexts, from foraging and nest relocation to-in the case of slavemaking species-slave raiding. Here, we elucidate the transcriptomic basis of scouting, tandem-leading and tandem-following behaviours across two species with divergent lifestyles: the slavemaking Temnothorax americanus and its primary, nonparasitic host T. longispinosus. Analysis of gene expression data from brains revealed that only a small number of unique differentially expressed genes are responsible for scouting and tandem-running. Comparison of orthologous genes between T. americanus and T. longispinosus suggests that tandem-running is characterized by species-specific patterns of gene usage. However, within both species, tandem-leaders showed gene expression patterns median to those of scouts and tandem-followers, which was expected, as leaders can be recruited from either of the other two behavioural states. Most importantly, a number of differentially expressed behavioural genes were found, with functions relating to learning and memory formation in other social and nonsocial insects. This includes a number of up-regulated receptor genes such as a glutamate and dopamine receptor, as well as serine/threonine-protein phosphatases and kinases. Learning and memory genes were specifically up-regulated within scouts and tandem-followers, not only reinforcing previous behavioural studies into how Temnothorax navigate novel environments and share information, but also providing insight into the molecular underpinnings of teaching and learning within social insects.
Collapse
Affiliation(s)
- Austin Alleman
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Marah Stoldt
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Barbara Feldmeyer
- Senckenberg Biodiversity and Climate Research Centre, Senckenberg Gesellschaft für Naturforschung, Frankfurt am Main, Germany
| | - Susanne Foitzik
- Institute of Organismic and Molecular Evolution, Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
24
|
Triacylglycerol Metabolism in Drosophila melanogaster. Genetics 2019; 210:1163-1184. [PMID: 30523167 DOI: 10.1534/genetics.118.301583] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2018] [Accepted: 09/11/2018] [Indexed: 12/11/2022] Open
Abstract
Triacylglycerol (TAG) is the most important caloric source with respect to energy homeostasis in animals. In addition to its evolutionarily conserved importance as an energy source, TAG turnover is crucial to the metabolism of structural and signaling lipids. These neutral lipids are also key players in development and disease. Here, we review the metabolism of TAG in the Drosophila model system. Recently, the fruit fly has attracted renewed attention in research due to the unique experimental approaches it affords in studying the tissue-autonomous and interorgan regulation of lipid metabolism in vivo Following an overview of the systemic control of fly body fat stores, we will cover lipid anabolic, enzymatic, and regulatory processes, which begin with the dietary lipid breakdown and de novo lipogenesis that results in lipid droplet storage. Next, we focus on lipolytic processes, which mobilize storage TAG to make it metabolically accessible as either an energy source or as a building block for biosynthesis of other lipid classes. Since the buildup and breakdown of fat involves various organs, we highlight avenues of lipid transport, which are at the heart of functional integration of organismic lipid metabolism. Finally, we draw attention to some "missing links" in basic neutral lipid metabolism and conclude with a perspective on how fly research can be exploited to study functional metabolic roles of diverse lipids.
Collapse
|
25
|
Aguilar G, Matsuda S, Vigano MA, Affolter M. Using Nanobodies to Study Protein Function in Developing Organisms. Antibodies (Basel) 2019; 8:E16. [PMID: 31544822 PMCID: PMC6640693 DOI: 10.3390/antib8010016] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 01/30/2019] [Accepted: 02/01/2019] [Indexed: 12/28/2022] Open
Abstract
Polyclonal and monoclonal antibodies have been invaluable tools to study proteins over the past decades. While indispensable for most biological studies including developmental biology, antibodies have been used mostly in fixed tissues or as binding reagents in the extracellular milieu. For functional studies and for clinical applications, antibodies have been functionalized by covalently fusing them to heterologous partners (i.e., chemicals, proteins or other moieties). Such functionalized antibodies have been less widely used in developmental biology studies. In the past few years, the discovery and application of small functional binding fragments derived from single-chain antibodies, so-called nanobodies, has resulted in novel approaches to study proteins during the development of multicellular animals in vivo. Expression of functionalized nanobody fusions from integrated transgenes allows manipulating proteins of interest in the extracellular and the intracellular milieu in a tissue- and time-dependent manner in an unprecedented manner. Here, we describe how nanobodies have been used in the field of developmental biology and look into the future to imagine how else nanobody-based reagents could be further developed to study the proteome in living organisms.
Collapse
Affiliation(s)
- Gustavo Aguilar
- Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland.
| | - Shinya Matsuda
- Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland.
| | - M Alessandra Vigano
- Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland.
| | - Markus Affolter
- Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland.
| |
Collapse
|
26
|
Li-Kroeger D, Kanca O, Lee PT, Cowan S, Lee MT, Jaiswal M, Salazar JL, He Y, Zuo Z, Bellen HJ. An expanded toolkit for gene tagging based on MiMIC and scarless CRISPR tagging in Drosophila. eLife 2018; 7:e38709. [PMID: 30091705 PMCID: PMC6095692 DOI: 10.7554/elife.38709] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023] Open
Abstract
We generated two new genetic tools to efficiently tag genes in Drosophila. The first, Double Header (DH) utilizes intronic MiMIC/CRIMIC insertions to generate artificial exons for GFP mediated protein trapping or T2A-GAL4 gene trapping in vivo based on Cre recombinase to avoid embryo injections. DH significantly increases integration efficiency compared to previous strategies and faithfully reports the expression pattern of genes and proteins. The second technique targets genes lacking coding introns using a two-step cassette exchange. First, we replace the endogenous gene with an excisable compact dominant marker using CRISPR making a null allele. Second, the insertion is replaced with a protein::tag cassette. This sequential manipulation allows the generation of numerous tagged alleles or insertion of other DNA fragments that facilitates multiple downstream applications. Both techniques allow precise gene manipulation and facilitate detection of gene expression, protein localization and assessment of protein function, as well as numerous other applications.
Collapse
Affiliation(s)
- David Li-Kroeger
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Oguz Kanca
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Pei-Tseng Lee
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Sierra Cowan
- Department of Biochemistry and Cell BiologyRice University HoustonHoustonUnited States
| | - Michael T Lee
- Department of Biochemistry and Cell BiologyRice University HoustonHoustonUnited States
| | - Manish Jaiswal
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Howard Hughes Medical InstituteBaylor College of MedicineHoustonUnited States
| | - Jose Luis Salazar
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Program in Developmental BiologyBaylor College of MedicineHoustonUnited States
| | - Yuchun He
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Howard Hughes Medical InstituteBaylor College of MedicineHoustonUnited States
| | - Zhongyuan Zuo
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
| | - Hugo J Bellen
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonUnited States
- Howard Hughes Medical InstituteBaylor College of MedicineHoustonUnited States
- Program in Developmental BiologyBaylor College of MedicineHoustonUnited States
- Department of NeuroscienceBaylor College of MedicineHoustonUnited States
- Jan and Dan Duncan Neurological Research InstituteHoustonUnited States
| |
Collapse
|
27
|
Lee PT, Zirin J, Kanca O, Lin WW, Schulze KL, Li-Kroeger D, Tao R, Devereaux C, Hu Y, Chung V, Fang Y, He Y, Pan H, Ge M, Zuo Z, Housden BE, Mohr SE, Yamamoto S, Levis RW, Spradling AC, Perrimon N, Bellen HJ. A gene-specific T2A-GAL4 library for Drosophila. eLife 2018; 7:35574. [PMID: 29565247 PMCID: PMC5898912 DOI: 10.7554/elife.35574] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/16/2018] [Indexed: 12/18/2022] Open
Abstract
We generated a library of ~1000 Drosophila stocks in which we inserted a construct in the intron of genes allowing expression of GAL4 under control of endogenous promoters while arresting transcription with a polyadenylation signal 3’ of the GAL4. This allows numerous applications. First, ~90% of insertions in essential genes cause a severe loss-of-function phenotype, an effective way to mutagenize genes. Interestingly, 12/14 chromosomes engineered through CRISPR do not carry second-site lethal mutations. Second, 26/36 (70%) of lethal insertions tested are rescued with a single UAS-cDNA construct. Third, loss-of-function phenotypes associated with many GAL4 insertions can be reverted by excision with UAS-flippase. Fourth, GAL4 driven UAS-GFP/RFP reports tissue and cell-type specificity of gene expression with high sensitivity. We report the expression of hundreds of genes not previously reported. Finally, inserted cassettes can be replaced with GFP or any DNA. These stocks comprise a powerful resource for assessing gene function. Determining what role newly discovered genes play in the body is an important part of genetics. This task requires a lot of extra information about each gene, such as the specific cells where the gene is active, or what happens when the gene is deleted. To answer these questions, researchers need tools and methods to manipulate genes within a living organism. The fruit fly Drosophila is useful for such experiments because a toolbox of genetic techniques is already available. Gene editing in fruit flies allows small pieces of genetic information to be removed from or added to anywhere in the animal’s DNA. Another tool, known as GAL4-UAS, is a two-part system used to study gene activity. The GAL4 component is a protein that switches on genes. GAL4 alone does very little in Drosophila cells because it only recognizes a DNA sequence called UAS. However, if a GAL4-producing cell is also engineered to contain a UAS-controlled gene, GAL4 will switch the gene on. Lee et al. used gene editing to insert a small piece of DNA, containing the GAL4 sequence followed by a ‘stop’ signal, into many different fly genes. The insertion made the cells where each gene was normally active produce GAL4, but – thanks to the stop signal – rendered the rest of the original gene non-functional. This effectively deleted the proteins encoded by each gene, giving information about the biological processes they normally control. Lee et al. went on to use their insertion approach to make a Drosophila genetic library. This is a collection of around 1,000 different strains of fly, each carrying the GAL4/stop combination in a single gene. The library allows any gene in the collection to be studied in detail simply by combining the GAL4 with different UAS-controlled genetic tools. For example, introducing a UAS-controlled marker would pinpoint where in the body the original gene was active. Alternatively, adding UAS-controlled human versions of the gene would create humanized flies, which are a valuable tool to study potential disease-causing genes in humans. This Drosophila library is a resource that contributes new experimental tools to fly genetics. Insights gained from flies can also be applied to more complex animals like humans, especially since around 65% of genes are similar across humans and Drosophila. As such, Lee et al. hope that this resource will help other researchers shed new light on the role of many different genes in health and disease.
Collapse
Affiliation(s)
- Pei-Tseng Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Jonathan Zirin
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Oguz Kanca
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Wen-Wen Lin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Karen L Schulze
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - David Li-Kroeger
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Rong Tao
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Colby Devereaux
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Yanhui Hu
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Verena Chung
- Department of Genetics, Harvard Medical School, Boston, United States
| | - Ying Fang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Yuchun He
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Hongling Pan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States
| | - Ming Ge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States
| | | | - Stephanie E Mohr
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Shinya Yamamoto
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States
| | - Robert W Levis
- Department of Embryology, Howard Hughes Medical Institute, Carnegie Institution for Science, Baltimore, United States
| | - Allan C Spradling
- Department of Embryology, Howard Hughes Medical Institute, Carnegie Institution for Science, Baltimore, United States
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, United States.,Howard Hughes Medical Institute, Harvard Medical School, Boston, United States
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, United States.,Program in Developmental Biology, Baylor College of Medicine, Houston, United States.,Department of Neuroscience, Baylor College of Medicine, Houston, United States
| |
Collapse
|