1
|
Ahmad M, Wu S, Luo S, Shi W, Guo X, Cao Y, Perrimon N, He L. Dietary amino acids promote glucagon-like hormone release to generate global calcium waves in adipose tissues in Drosophila. Nat Commun 2025; 16:247. [PMID: 39747032 PMCID: PMC11696257 DOI: 10.1038/s41467-024-55371-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Propagation of intercellular calcium waves through tissues has been found to coordinate different multicellular responses. Nevertheless, our understanding of how calcium waves operate remains limited. In this study, we explore the real-time dynamics of intercellular calcium waves in Drosophila adipose tissues. We identify Adipokinetic Hormone (AKH), the fly functional homolog of glucagon, as the key factor driving Ca2+ activities in adipose tissue. We find that AKH, which is released into the hemolymph from the AKH-producing neurosecretory cells, stimulates calcium waves in the larval fat by a previously unrecognized gap-junction-independent mechanism to promote lipolysis. In the adult fat body, however, gap-junction-dependent intercellular calcium waves are triggered by a presumably uniformly diffused AKH. Additionally, we discover that amino acids activate the AKH-producing neurosecretory cells, leading to increased intracellular Ca2+ and AKH secretion. Altogether, we show that dietary amino acids regulate the AKH release from the AKH-producing neurosecretory cells in the brain, which subsequently stimulates gap-junction-independent intercellular calcium waves in adipose tissue, enhancing lipid metabolism.
Collapse
Affiliation(s)
- Muhammad Ahmad
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Shang Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shengyao Luo
- Yuanpei College, Peking University, Beijing, China
| | - Wenjia Shi
- Department of Applied Physics, Xi'an University of Technology, Xi'an, Shaanxi, China
| | - Xuan Guo
- Life Science Institute, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yuansheng Cao
- Department of Physics, Tsinghua University, Beijing, China.
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| | - Li He
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
2
|
Ho KYL, Ou AYJ, Samuelson N, Tanentzapf G. Novel features of Drosophila hematopoiesis uncovered by long-term live imaging. Dev Biol 2025; 517:286-300. [PMID: 39536928 DOI: 10.1016/j.ydbio.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/25/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024]
Abstract
Stem cells are subject to continuous regulation to ensure that the correct balance between stem cell differentiation and self-renewal is maintained. The dynamic and ongoing nature of stem cell regulation, as well as the complex signaling microenvironment in which stem cells are typically found, means that studying them in their endogenous environment in real time has multiple advantages over static fixed-sample approaches. We recently described a method for long-term, ex-vivo, live imaging of the blood progenitors in the Drosophila larval hematopoietic organ, the Lymph Gland (LG). This methodology has allowed us to analyze multiple aspects of fly hematopoiesis, in real time, in a manner that could not be carried out previously. Here, we describe novel insights derived from our quantitative live imaging approach. These insights include: the identification of extensive filopodia in the progenitors and description of their morphology and dynamics; visualization and quantitative analysis of JAK/STAT signaling in progenitors by the simultaneous tracking of thousands of vesicles containing internalized Domeless receptors; quantitative analysis of the location, morphology, and dynamics of mitochondria in blood progenitors; long-term tracking of patterns of cell division and migration of mature blood cell in the LG; long-term tracking of multiple cell behaviors in the distal committed progenitors; analysis of Ca2+ signaling of blood progenitors in the secondary lobes of the LG. Together, these observations illustrate the power of imaging fly hematopoiesis in real time and identify many previously undescribed processes and behaviors in the LG that are likely to play important roles in the regulation of progenitor differentiation and self-renewal.
Collapse
Affiliation(s)
- Kevin Y L Ho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Annie Y J Ou
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada; School of Kinesiology, University of British Columbia, Vancouver, V6T 1Z1, Canada; Laboratory of Molecular Immunology, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo, 113-0032, Japan
| | - Nicholas Samuelson
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, V6T 1Z3, Canada.
| |
Collapse
|
3
|
Penfield L, Montell DJ. Nuclear lamin facilitates collective border cell invasion into confined spaces in vivo. J Cell Biol 2023; 222:e202212101. [PMID: 37695420 PMCID: PMC10494525 DOI: 10.1083/jcb.202212101] [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: 12/20/2022] [Revised: 06/05/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Abstract
Cells migrate collectively through confined environments during development and cancer metastasis. The nucleus, a stiff organelle, impedes single cells from squeezing into narrow channels within artificial environments. However, how nuclei affect collective migration into compact tissues is unknown. Here, we use border cells in the fly ovary to study nuclear dynamics in collective, confined in vivo migration. Border cells delaminate from the follicular epithelium and squeeze into tiny spaces between cells called nurse cells. The lead cell nucleus transiently deforms within the lead cell protrusion, which then widens. The nuclei of follower cells deform less. Depletion of the Drosophila B-type lamin, Lam, compromises nuclear integrity, hinders expansion of leading protrusions, and impedes border cell movement. In wildtype, cortical myosin II accumulates behind the nucleus and pushes it into the protrusion, whereas in Lam-depleted cells, myosin accumulates but does not move the nucleus. These data suggest that the nucleus stabilizes lead cell protrusions, helping to wedge open spaces between nurse cells.
Collapse
Affiliation(s)
- Lauren Penfield
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Denise J. Montell
- Department of Molecular, Cellular, and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
4
|
Das M, Cheng D, Matzat T, Auld VJ. Innexin-Mediated Adhesion between Glia Is Required for Axon Ensheathment in the Peripheral Nervous System. J Neurosci 2023; 43:2260-2276. [PMID: 36801823 PMCID: PMC10072304 DOI: 10.1523/jneurosci.1323-22.2023] [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: 07/05/2022] [Revised: 01/03/2023] [Accepted: 02/09/2023] [Indexed: 02/19/2023] Open
Abstract
Glia are essential to protecting and enabling nervous system function and a key glial function is the formation of the glial sheath around peripheral axons. Each peripheral nerve in the Drosophila larva is ensheathed by three glial layers, which structurally support and insulate the peripheral axons. How peripheral glia communicate with each other and between layers is not well established and we investigated the role of Innexins in mediating glial function in the Drosophila periphery. Of the eight Drosophila Innexins, we found two (Inx1 and Inx2) are important for peripheral glia development. In particular loss of Inx1 and Inx2 resulted in defects in the wrapping glia leading to disruption of the glia wrap. Of interest loss of Inx2 in the subperineurial glia also resulted in defects in the neighboring wrapping glia. Inx plaques were observed between the subperineurial glia and the wrapping glia suggesting that gap junctions link these two glial cell types. We found Inx2 is key to Ca2+ pulses in the peripheral subperineurial glia but not in the wrapping glia, and we found no evidence of gap junction communication between subperineurial and wrapping glia. Rather we have clear evidence that Inx2 plays an adhesive and channel-independent role between the subperineurial and wrapping glia to ensure the integrity of the glial wrap.SIGNIFICANCE STATEMENT Gap junctions are critical for glia communication and formation of myelin in myelinating glia. However, the role of gap junctions in non-myelinating glia is not well studied, yet non-myelinating glia are critical for peripheral nerve function. We found the Innexin gap junction proteins are present between different classes of peripheral glia in Drosophila. Here Innexins form junctions to facilitate adhesion between the different glia but do so in a channel-independent manner. Loss of adhesion leads to disruption of the glial wrap around axons and leads to fragmentation of the wrapping glia membranes. Our work points to an important role for gap junction proteins in mediating insulation by non-myelinating glia.
Collapse
Affiliation(s)
- Mriga Das
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Duo Cheng
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Till Matzat
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| | - Vanessa J Auld
- Department of Zoology, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
5
|
Bioelectric regulation of intestinal stem cells. Trends Cell Biol 2022:S0962-8924(22)00234-3. [PMID: 36396487 PMCID: PMC10183058 DOI: 10.1016/j.tcb.2022.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 11/17/2022]
Abstract
Proper regulation of ion balance across the intestinal epithelium is essential for physiological functions, while ion imbalance causes intestinal disorders with dire health consequences. Ion channels, pumps, and exchangers are vital for regulating ion movements (i.e., bioelectric currents) that control epithelial absorption and secretion. Recent in vivo studies used the Drosophila gut to identify conserved pathways that link regulators of Ca2+, Na+ and Cl- with intestinal stem cell (ISC) proliferation. These studies laid a foundation for using the Drosophila gut to identify conserved proliferative responses triggered by bioelectric regulators. Here, we review these studies, discuss their significance, as well as the advantages of using Drosophila to unravel conserved bioelectrically induced molecular pathways in the intestinal epithelium under physiological, pathophysiological, and regenerative conditions.
Collapse
|
6
|
Mallart C, Chalvet F, Netter S, Torres AY, Poidevin M, Montagne J, Pret AM, Malartre M. E-cadherin acts as a positive regulator of the JAK-STAT signaling pathway during Drosophila oogenesis. Front Cell Dev Biol 2022; 10:886312. [PMID: 36120588 PMCID: PMC9473917 DOI: 10.3389/fcell.2022.886312] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 07/14/2022] [Indexed: 11/15/2022] Open
Abstract
The JAK-STAT pathway is evolutionary conserved. The simplicity of this signaling in Drosophila, due to the limited redundancy between pathway components, makes it an ideal model for investigation. In the Drosophila follicular epithelium, highly stereotyped functions of JAK-STAT signaling have been well characterized, but how signaling activity is regulated precisely to allow the different outcomes is not well understood. In this tissue, the ligand is secreted by the polar cells positioned at each follicle extremity, thus generating a gradient of JAK-STAT activity in adjacent cells. One way to control the delivered quantity of ligand is by regulating the number of polar cells, which is reduced by apoptosis to exactly two at each pole by mid-oogenesis. Hence, JAK-STAT activity is described as symmetrical between follicle anterior and posterior regions. Here, we show that JAK-STAT signaling activity is actually highly dynamic, resulting in asymmetry between poles by mid-oogenesis. Interestingly, we found similar temporal dynamics at follicle poles in the accumulation of the adherens junction E-cadherin protein. Remarkably, E-cadherin and JAK-STAT signaling not only display patterning overlaps but also share functions during oogenesis. In particular, we show that E-cadherin, like JAK-STAT signaling, regulates polar cell apoptosis non-cell-autonomously from follicle cells. Finally, our work reveals that E-cadherin is required for optimal JAK-STAT activity throughout oogenesis and that E-cadherin and Stat92E, the transcription factor of the pathway, form part of a physical complex in follicle cells. Taken together, our study establishes E-cadherin as a new positive regulator of JAK-STAT signaling during oogenesis.
Collapse
Affiliation(s)
- Charlotte Mallart
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Fabienne Chalvet
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Sophie Netter
- Institute for Integrative Biology of the Cell (I2BC), UVSQ, CEA, CNRS, Université Paris-Saclay, Gif- sur-Yvette, France
| | - Alba Yurani Torres
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Mickael Poidevin
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Jacques Montagne
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Anne-Marie Pret
- Institute for Integrative Biology of the Cell (I2BC), UVSQ, CEA, CNRS, Université Paris-Saclay, Gif- sur-Yvette, France
| | - Marianne Malartre
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette, France
- *Correspondence: Marianne Malartre,
| |
Collapse
|
7
|
Ho KYL, Khadilkar RJ, Carr RL, Tanentzapf G. A gap-junction-mediated, calcium-signaling network controls blood progenitor fate decisions in hematopoiesis. Curr Biol 2021; 31:4697-4712.e6. [PMID: 34480855 DOI: 10.1016/j.cub.2021.08.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 05/28/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022]
Abstract
Stem cell homeostasis requires coordinated fate decisions among stem cells that are often widely distributed within a tissue at varying distances from their stem cell niche. This requires a mechanism to ensure robust fate decisions within a population of stem cells. Here, we show that, in the Drosophila hematopoietic organ, the lymph gland (LG), gap junctions form a network that coordinates fate decisions between blood progenitors. Using live imaging of calcium signaling in intact LGs, we find that blood progenitors are connected through a signaling network. Blocking gap junction function disrupts this network, alters the pattern of encoded calcium signals, and leads to loss of progenitors and precocious blood cell differentiation. Ectopic and uniform activation of the calcium-signaling mediator CaMKII restores progenitor homeostasis when gap junctions are disrupted. Overall, these data show that gap junctions equilibrate cell signals between blood progenitors to coordinate fate decisions and maintain hematopoietic homeostasis.
Collapse
Affiliation(s)
- Kevin Y L Ho
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Rohan J Khadilkar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; Advanced Centre for Treatment, Research and Education in Cancer-Tata Memorial Centre (ACTREC-TMC), Kharghar, Navi Mumbai, Maharashtra 410210, India
| | - Rosalyn L Carr
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Guy Tanentzapf
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada.
| |
Collapse
|
8
|
Neves JH, Rezende-Teixeira P, Palomino NB, Machado-Santelli GM. Molecular and morphological approach to study the innexin gap junctions in Rhynchosciara americana. Open Biol 2021; 11:210224. [PMID: 34753320 PMCID: PMC8580445 DOI: 10.1098/rsob.210224] [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] [Indexed: 01/04/2023] Open
Abstract
Gap junctions mediate communication between adjacent cells and are fundamental to the development and homeostasis in multicellular organisms. In invertebrates, gap junctions are formed by transmembrane proteins called innexins. Gap junctions allow the passage of small molecules through an intercellular channel, between a cell and another adjacent cell. The dipteran Rhynchosciara americana has contributed to studying the biology of invertebrates and the study of the interaction and regulation of genes during biological development. Therefore, this paper aimed to study the R. americana innexin-2 by molecular characterization, analysis of the expression profile and cellular localization. The molecular characterization results confirm that the message is from a gap junction protein and analysis of the expression and cellular localization profile shows that innexin-2 can participate in many physiological processes during the development of R. americana.
Collapse
Affiliation(s)
- Jorge Henrique Neves
- Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 1524 – sala 307, São Paulo, SP, Brazil
| | - Paula Rezende-Teixeira
- Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 1524 – sala 307, São Paulo, SP, Brazil
| | - Natalia Bazan Palomino
- Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 1524 – sala 307, São Paulo, SP, Brazil
| | - Glaucia Maria Machado-Santelli
- Institute of Biomedical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 1524 – sala 307, São Paulo, SP, Brazil
| |
Collapse
|
9
|
Huang YC, Chen KH, Chen YY, Tsao LH, Yeh TH, Chen YC, Wu PY, Wang TW, Yu JY. βPS-Integrin acts downstream of Innexin 2 in modulating stretched cell morphogenesis in the Drosophila ovary. G3-GENES GENOMES GENETICS 2021; 11:6310741. [PMID: 34544125 PMCID: PMC8496311 DOI: 10.1093/g3journal/jkab215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/14/2021] [Indexed: 11/25/2022]
Abstract
During oogenesis, a group of specialized follicle cells, known as stretched cells (StCs), flatten drastically from cuboidal to squamous shape. While morphogenesis of epithelia is critical for organogenesis, genes and signaling pathways involved in this process remain to be revealed. In addition to formation of gap junctions for intercellular exchange of small molecules, gap junction proteins form channels or act as adaptor proteins to regulate various cellular behaviors. In invertebrates, gap junction proteins are Innexins. Knockdown of Innexin 2 but not other Innexins expressed in follicle cells attenuates StC morphogenesis. Interestingly, blocking of gap junctions with an inhibitor carbenoxolone does not affect StC morphogenesis, suggesting that Innexin 2 might control StCs flattening in a gap-junction-independent manner. An excessive level of βPS-Integrin encoded by myospheroid is detected in Innexin 2 mutant cells specifically during StC morphogenesis. Simultaneous knockdown of Innexin 2 and myospheroid partially rescues the morphogenetic defect resulted from Innexin 2 knockdown. Furthermore, reduction of βPS-Integrin is sufficient to induce early StCs flattening. Taken together, our data suggest that βPS-Integrin acts downstream of Innexin 2 in modulating StCs morphogenesis.
Collapse
Affiliation(s)
- Yi-Chia Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Kuan-Han Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Yang Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Liang-Hsuan Tsao
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Tsung-Han Yeh
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Yu-Chia Chen
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ping-Yen Wu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Tsu-Wei Wang
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan
| | - Jenn-Yah Yu
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.,Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| |
Collapse
|
10
|
Germline soma communication mediated by gap junction proteins regulates epithelial morphogenesis. PLoS Genet 2021; 17:e1009685. [PMID: 34343194 PMCID: PMC8330916 DOI: 10.1371/journal.pgen.1009685] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/28/2021] [Indexed: 01/22/2023] Open
Abstract
Gap junction (GJ) proteins, the primary constituents of GJ channels, are conserved determinants of patterning. Canonically, a GJ channel, made up of two hemi-channels contributed by the neighboring cells, facilitates transport of metabolites/ions. Here we demonstrate the involvement of GJ proteins during cuboidal to squamous epithelial transition displayed by the anterior follicle cells (AFCs) from Drosophila ovaries. Somatically derived AFCs stretch and flatten when the adjacent germline cells start increasing in size. GJ proteins, Innexin2 (Inx2) and Innexin4 (Inx4), functioning in the AFCs and germline respectively, promote the shape transformation by modulating calcium levels in the AFCs. Our observations suggest that alterations in calcium flux potentiate STAT activity to influence actomyosin-based cytoskeleton, possibly resulting in disassembly of adherens junctions. Our data have uncovered sequential molecular events underlying the cuboidal to squamous shape transition and offer unique insight into how GJ proteins expressed in the neighboring cells contribute to morphogenetic processes. Shape transitions between different subtypes of epithelial cells i.e., cuboidal, squamous and columnar are ubiquitous and are essential during organogenesis across animal kingdom. We demonstrate that heteromeric combination of gap junction proteins, Drosophila Innexin2 and Drosophila Innexin 4 (also known as Zero population growth or Zpg), expressed in the soma and germline of fly egg respectively, mediates the shape transition of cuboidal follicle cells to squamous fate. Interestingly, the two gap junction proteins likely participate as constituents of a calcium channel. Further, we show that somatic follicle cells and germline nurse cells communicate through calcium fluxes that activates STAT in the follicle cells. Activated STAT modulates the levels/ activity of junctional complexes thus aiding shape transition of cuboidal cells to squamous fate. These findings provide novel insights into how communication between different cell types with distinct origins achieve shape transitions essential for proper organ assemblies.
Collapse
|
11
|
Li C, Shi L, Peng C, Yu G, Zhang Y, Du Z. Lead-induced cardiomyocytes apoptosis by inhibiting gap junction intercellular communication via autophagy activation. Chem Biol Interact 2020; 337:109331. [PMID: 33242459 DOI: 10.1016/j.cbi.2020.109331] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 11/01/2020] [Accepted: 11/20/2020] [Indexed: 12/15/2022]
Abstract
Lead (Pb) is one of the most common heavy metal contaminants in the environment. Pb can cause pathophysiological changes in several organ systems, including the cardiovascular system, but the molecular mechanism remains elusive. The study aimed to study the effects of Pb on Gap junction intercellular communication (GJIC) and its role in Pb-induced apoptosis. The present study aims to determine whether Pb-induced autophagy promotes apoptosis of rat cardiac myocytes (H9c2 cells) by downregulating GJIC using CCK-8 Kit, scrape loading/dye transfer assay, Annexin V/PI assays, Western blot analysis and double-immunofluorescence experiments. The results showed that Pb elicited cytotoxicity in a time- and concentration-dependent manner and led to increased apoptosis in a concentration-dependent manner in H9c2 cells. Pb also reduced GJIC in H9c2 cells in a concentration-dependent manner through the downregulation of connexin (Cx) 43. Inhibition of gap junctions by gap junction blocker carbenoxolone disodium (CBX) resulted in increased apoptosis. Furthermore, Pb increased autophagy in a concentration-dependent manner in H9c2 cells, decreasing the distribution of Cx43 on the cell membrane, and targeted Cx43 to autophagosome via light chain 3 (LC3). However, autophagy inhibitor 3-Methyladenine (3-MA) can slow down the downregulation of Cx43 induced by Pb in H9c2 cells. In conclusion, our results provide evidence that Pb-decreased GJIC promotes apoptosis in cardiomyocytes. This is probably because of the fact that Pb-induced autophagy exacerbates GJIC inhibition and downregulation of Cx43.
Collapse
Affiliation(s)
- Chao Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China; Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China
| | - Liang Shi
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China
| | - Cheng Peng
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China; Queensland Alliance for Environmental Health Sciences (QAEHS), The University of Queensland, Brisbane, 4108, Queensland, Australia
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China
| | - Yanshu Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, Hebei, China; Laboratory Animal Center, North China University of Science and Technology, Tangshan, 063210, Hebei, China.
| | - Zhongjun Du
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Ji'nan, 250062, Shandong, China.
| |
Collapse
|
12
|
Miao G, Godt D, Montell DJ. Integration of Migratory Cells into a New Site In Vivo Requires Channel-Independent Functions of Innexins on Microtubules. Dev Cell 2020; 54:501-515.e9. [PMID: 32668209 DOI: 10.1016/j.devcel.2020.06.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/03/2020] [Accepted: 06/19/2020] [Indexed: 12/31/2022]
Abstract
During embryonic development and cancer metastasis, migratory cells must establish stable connections with new partners at their destinations. Here, we establish the Drosophila border cells as a model for this multistep process. During oogenesis, border cells delaminate from the follicular epithelium and migrate. When they reach their target, the oocyte, they undergo a stereotypical series of steps to adhere to it, then connect with another migrating epithelium. We identify gap-junction-forming innexin proteins as critical. Surprisingly, the channel function is dispensable. Instead, Innexins 2 and 3 function within the border cells, and Innexin 4 functions within the germline, to regulate microtubules. The microtubule-dependent border cell-oocyte interaction is essential to brace the cells against external morphogenetic forces. Thus, we establish an experimental model and use genetic, thermogenetic, and live-imaging approaches to uncover the contributions of Innexins and microtubules to a cell-biological process important in development and cancer.
Collapse
Affiliation(s)
- Guangxia Miao
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA 93106, USA
| | - Dorothea Godt
- Department of Cell and Systems Biology, University of Toronto, 25 Harbord Street, Toronto, ON M5S 3G5, Canada
| | - Denise J Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, Santa Barbara, CA 93106, USA.
| |
Collapse
|
13
|
Schotthöfer SK, Bohrmann J. Analysing bioelectrical phenomena in the Drosophila ovary with genetic tools: tissue-specific expression of sensors for membrane potential and intracellular pH, and RNAi-knockdown of mechanisms involved in ion exchange. BMC DEVELOPMENTAL BIOLOGY 2020; 20:15. [PMID: 32635900 PMCID: PMC7341674 DOI: 10.1186/s12861-020-00220-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/10/2020] [Indexed: 01/16/2023]
Abstract
Background Changes in transcellular bioelectrical patterns are known to play important roles during developmental and regenerative processes. The Drosophila follicular epithelium has proven to be an appropriate model system for studying the mechanisms by which bioelectrical signals emerge and act. Fluorescent indicator dyes in combination with various inhibitors of ion-transport mechanisms have been used to investigate the generation of membrane potentials (Vmem) and intracellular pH (pHi). Both parameters as well as their anteroposterior and dorsoventral gradients were affected by the inhibitors which, in addition, led to alterations of microfilament and microtubule patterns equivalent to those observed during follicle-cell differentiation. Results We expressed two genetically-encoded fluorescent sensors for Vmem and pHi, ArcLight and pHluorin-Moesin, in the follicular epithelium of Drosophila. By means of the respective inhibitors, we obtained comparable effects on Vmem and/or pHi as previously described for Vmem- and pHi-sensitive fluorescent dyes. In a RNAi-knockdown screen, five genes of ion-transport mechanisms and gap-junction subunits were identified exerting influence on ovary development and/or oogenesis. Loss of ovaries or small ovaries were the results of soma knockdowns of the innexins inx1 and inx3, and of the DEG/ENaC family member ripped pocket (rpk). Germline knockdown of rpk also resulted in smaller ovaries. Soma knockdown of the V-ATPase-subunit vha55 caused size-reduced ovaries with degenerating follicles from stage 10A onward. In addition, soma knockdown of the open rectifier K+channel 1 (ork1) resulted in a characteristic round-egg phenotype with altered microfilament and microtubule organisation in the follicular epithelium. Conclusions The genetic tool box of Drosophila provides means for a refined and extended analysis of bioelectrical phenomena. Tissue-specifically expressed Vmem- and pHi-sensors exhibit some practical advantages compared to fluorescent indicator dyes. Their use confirms that the ion-transport mechanisms targeted by inhibitors play important roles in the generation of bioelectrical signals. Moreover, modulation of bioelectrical signals via RNAi-knockdown of genes coding for ion-transport mechanisms and gap-junction subunits exerts influence on crucial processes during ovary development and results in cytoskeletal changes and altered follicle shape. Thus, further evidence amounts for bioelectrical regulation of developmental processes via the control of both signalling pathways and cytoskeletal organisation.
Collapse
Affiliation(s)
- Susanne Katharina Schotthöfer
- RWTH Aachen University, Institut für Biologie II, Abt. Zoologie und Humanbiologie, Worringerweg 3, 52056, Aachen, Germany
| | - Johannes Bohrmann
- RWTH Aachen University, Institut für Biologie II, Abt. Zoologie und Humanbiologie, Worringerweg 3, 52056, Aachen, Germany.
| |
Collapse
|
14
|
Zhao S, Mehta AS, Zhao M. Biomedical applications of electrical stimulation. Cell Mol Life Sci 2020; 77:2681-2699. [PMID: 31974658 PMCID: PMC7954539 DOI: 10.1007/s00018-019-03446-1] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/12/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
This review provides a comprehensive overview on the biomedical applications of electrical stimulation (EStim). EStim has a wide range of direct effects on both biomolecules and cells. These effects have been exploited to facilitate proliferation and functional development of engineered tissue constructs for regenerative medicine applications. They have also been tested or used in clinics for pain mitigation, muscle rehabilitation, the treatment of motor/consciousness disorders, wound healing, and drug delivery. However, the research on fundamental mechanism of cellular response to EStim has fell behind its applications, which has hindered the full exploitation of the clinical potential of EStim. Moreover, despite the positive outcome from the in vitro and animal studies testing the efficacy of EStim, existing clinical trials failed to establish strong, conclusive supports for the therapeutic efficacy of EStim for most of the clinical applications mentioned above. Two potential directions of future research to improve the clinical utility of EStim are presented, including the optimization and standardization of the stimulation protocol and the development of more tissue-matching devices.
Collapse
Affiliation(s)
- Siwei Zhao
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, 985965 Nebraska Medical Center, Omaha, NE, 68198, USA.
- Department of Surgery, University of Nebraska Medical Center, Nebraska Medical Center 985965, Omaha, NE, 68198, USA.
| | - Abijeet Singh Mehta
- Department of Dermatology, University of California, Davis, CA, USA
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| | - Min Zhao
- Department of Dermatology, University of California, Davis, CA, USA
- Department of Ophthalmology & Vision Science, Institute for Regenerative Cures, Center for Neuroscience, University of California at Davis, School of Medicine, Suite 1630, Room 1617, 2921 Stockton Blvd., Sacramento, CA, 95817, USA
| |
Collapse
|
15
|
Jevitt A, Chatterjee D, Xie G, Wang XF, Otwell T, Huang YC, Deng WM. A single-cell atlas of adult Drosophila ovary identifies transcriptional programs and somatic cell lineage regulating oogenesis. PLoS Biol 2020; 18:e3000538. [PMID: 32339165 PMCID: PMC7205450 DOI: 10.1371/journal.pbio.3000538] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 05/07/2020] [Accepted: 03/27/2020] [Indexed: 12/28/2022] Open
Abstract
Oogenesis is a complex developmental process that involves spatiotemporally regulated coordination between the germline and supporting, somatic cell populations. This process has been modeled extensively using the Drosophila ovary. Although different ovarian cell types have been identified through traditional means, the large-scale expression profiles underlying each cell type remain unknown. Using single-cell RNA sequencing technology, we have built a transcriptomic data set for the adult Drosophila ovary and connected tissues. Using this data set, we identified the transcriptional trajectory of the entire follicle-cell population over the course of their development from stem cells to the oogenesis-to-ovulation transition. We further identify expression patterns during essential developmental events that take place in somatic and germline cell types such as differentiation, cell-cycle switching, migration, symmetry breaking, nurse-cell engulfment, egg-shell formation, and corpus luteum signaling. Extensive experimental validation of unique expression patterns in both ovarian and nearby, nonovarian cells also led to the identification of many new cell type-and stage-specific markers. The inclusion of several nearby tissue types in this data set also led to our identification of functional convergence in expression between distantly related cell types such as the immune-related genes that were similarly expressed in immune cells (hemocytes) and ovarian somatic cells (stretched cells) during their brief phagocytic role in nurse-cell engulfment. Taken together, these findings provide new insight into the temporal regulation of genes in a cell-type specific manner during oogenesis and begin to reveal the relatedness in expression between cell and tissues types.
Collapse
Affiliation(s)
- Allison Jevitt
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Deeptiman Chatterjee
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Gengqiang Xie
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Taylor Otwell
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| | - Wu-Min Deng
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
| |
Collapse
|
16
|
Peercy BE, Starz-Gaiano M. Clustered cell migration: Modeling the model system of Drosophila border cells. Semin Cell Dev Biol 2019; 100:167-176. [PMID: 31837934 DOI: 10.1016/j.semcdb.2019.11.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 11/11/2019] [Accepted: 11/15/2019] [Indexed: 01/19/2023]
Abstract
In diverse developmental contexts, certain cells must migrate to fulfill their roles. Many questions remain unanswered about the genetic and physical properties that govern cell migration. While the simplest case of a single cell moving alone has been well-studied, additional complexities arise in considering how cohorts of cells move together. Significant differences exist between models of collectively migrating cells. We explore the experimental model of migratory border cell clusters in Drosophila melanogaster egg chambers, which are amenable to direct observation and precise genetic manipulations. This system involves two special characteristics that are worthy of attention: border cell clusters contain a limited number of both migratory and non-migratory cells that require coordination, and they navigate through a heterogeneous three-dimensional microenvironment. First, we review how clusters of motile border cells are specified and guided in their migration by chemical signals and the physical impact of adjacent tissue interactions. In the second part, we examine questions around the 3D structure of the motile cluster and surrounding microenvironment in understanding the limits to cluster size and speed of movement through the egg chamber. Mathematical models have identified sufficient gene regulatory networks for specification, the key forces that capture emergent behaviors observed in vivo, the minimal regulatory topologies for signaling, and the distribution of key signaling cues that direct cell behaviors. This interdisciplinary approach to studying border cells is likely to reveal governing principles that apply to different types of cell migration events.
Collapse
Affiliation(s)
- Bradford E Peercy
- Department of Mathematics and Statistics, UMBC, Baltimore, MD 21250, United States.
| | | |
Collapse
|
17
|
Voelker L, Upadhyaya B, Ferkey DM, Woldemariam S, L’Etoile ND, Rabinowitch I, Bai J. INX-18 and INX-19 play distinct roles in electrical synapses that modulate aversive behavior in Caenorhabditis elegans. PLoS Genet 2019; 15:e1008341. [PMID: 31658255 PMCID: PMC6837551 DOI: 10.1371/journal.pgen.1008341] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 11/07/2019] [Accepted: 10/04/2019] [Indexed: 12/23/2022] Open
Abstract
In order to respond to changing environments and fluctuations in internal states, animals adjust their behavior through diverse neuromodulatory mechanisms. In this study we show that electrical synapses between the ASH primary quinine-detecting sensory neurons and the neighboring ASK neurons are required for modulating the aversive response to the bitter tastant quinine in C. elegans. Mutant worms that lack the electrical synapse proteins INX-18 and INX-19 become hypersensitive to dilute quinine. Cell-specific rescue experiments indicate that inx-18 operates in ASK while inx-19 is required in both ASK and ASH for proper quinine sensitivity. Imaging analyses find that INX-19 in ASK and ASH localizes to the same regions in the nerve ring, suggesting that both sides of ASK-ASH electrical synapses contain INX-19. While inx-18 and inx-19 mutant animals have a similar behavioral phenotype, several lines of evidence suggest the proteins encoded by these genes play different roles in modulating the aversive quinine response. First, INX-18 and INX-19 localize to different regions of the nerve ring, indicating that they are not present in the same synapses. Second, removing inx-18 disrupts the distribution of INX-19, while removing inx-19 does not alter INX-18 localization. Finally, by using a fluorescent cGMP reporter, we find that INX-18 and INX-19 have distinct roles in establishing cGMP levels in ASK and ASH. Together, these results demonstrate that electrical synapses containing INX-18 and INX-19 facilitate modulation of ASH nociceptive signaling. Our findings support the idea that a network of electrical synapses mediates cGMP exchange between neurons, enabling modulation of sensory responses and behavior. Animals are constantly adjusting their behavior to respond to changes in the environment or to their internal state. This behavior modulation is achieved by altering the activity of neurons and circuits through a variety of neuroplasticity mechanisms. Chemical synapses are known to impact neuroplasticity in several different ways, but the diversity of mechanisms by which electrical synapses contribute is still being investigated. Electrical synapses are specialized sites of connection between neurons where ions and small signaling molecules can pass directly from one cell to the next. By passing small molecules through electrical synapses, neurons may be able to modify the activity of their neighbors. In this study we identify two genes that contribute to electrical synapses between two sensory neurons in C. elegans. We show that these electrical synapses are crucial for proper modulation of sensory responses, as without them animals are overly responsive to an aversive stimulus. In addition to pinpointing their sites of action, we present evidence that they may be contributing to neuromodulation by facilitating passage of the small molecule cGMP between neurons. Our work provides evidence for a role of electrical synapses in regulating animal behavior.
Collapse
Affiliation(s)
- Lisa Voelker
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, United States of America
| | - Bishal Upadhyaya
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
| | - Denise M. Ferkey
- Department of Biological Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States of America
| | - Sarah Woldemariam
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, United States of America
| | - Noelle D. L’Etoile
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, United States of America
| | - Ithai Rabinowitch
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Department of Medical Neurobiology, Faculty of Medicine Hebrew, University of Jerusalem, Jerusalem, Israel
| | - Jihong Bai
- Basic Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States of America
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, United States of America
- * E-mail:
| |
Collapse
|
18
|
Guo X, Luo J, Wang H, Chen J. SERCA regulates collective cell migration by maintaining cytoplasmic Ca 2+ homeostasis. J Genet Genomics 2019; 46:451-454. [PMID: 31611171 DOI: 10.1016/j.jgg.2019.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Affiliation(s)
- Xuan Guo
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China
| | - Jun Luo
- College of Life Science, Shangrao Normal University, Shangrao, 334001, China
| | - Heng Wang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
| | - Jiong Chen
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animals for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, 210061, China.
| |
Collapse
|
19
|
Shyu WH, Lee WP, Chiang MH, Chang CC, Fu TF, Chiang HC, Wu T, Wu CL. Electrical synapses between mushroom body neurons are critical for consolidated memory retrieval in Drosophila. PLoS Genet 2019; 15:e1008153. [PMID: 31071084 PMCID: PMC6529013 DOI: 10.1371/journal.pgen.1008153] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/21/2019] [Accepted: 04/23/2019] [Indexed: 11/19/2022] Open
Abstract
Electrical synapses between neurons, also known as gap junctions, are direct cell membrane channels between adjacent neurons. Gap junctions play a role in the synchronization of neuronal network activity; however, their involvement in cognition has not been well characterized. Three-hour olfactory associative memory in Drosophila has two components: consolidated anesthesia-resistant memory (ARM) and labile anesthesia-sensitive memory (ASM). Here, we show that knockdown of the gap junction gene innexin5 (inx5) in mushroom body (MB) neurons disrupted ARM, while leaving ASM intact. Whole-mount brain immunohistochemistry indicated that INX5 protein was preferentially expressed in the somas, calyxes, and lobes regions of the MB neurons. Adult-stage-specific knockdown of inx5 in αβ neurons disrupted ARM, suggesting a specific requirement of INX5 in αβ neurons for ARM formation. Hyperpolarization of αβ neurons during memory retrieval by expressing an engineered halorhodopsin (eNpHR) also disrupted ARM. Administration of the gap junction blocker carbenoxolone (CBX) reduced the proportion of odor responsive αβ neurons to the training odor 3 hours after training. Finally, the α-branch-specific 3-hour ARM-specific memory trace was also diminished with CBX treatment and in inx5 knockdown flies. Altogether, our results suggest INX5 gap junction channels in αβ neurons for ARM retrieval and also provide a more detailed neuronal mechanism for consolidated memory in Drosophila.
Collapse
Affiliation(s)
- Wei-Huan Shyu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wang-Pao Lee
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Meng-Hsuan Chiang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Ching Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsai-Feng Fu
- Department of Applied Chemistry, National Chi Nan University, Nantou, Taiwan
| | - Hsueh-Cheng Chiang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tony Wu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chia-Lin Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Biochemistry, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Neurology, Chang Gung Memorial Hospital, Linkou, Taiwan
- * E-mail:
| |
Collapse
|
20
|
Brodskiy PA, Wu Q, Soundarrajan DK, Huizar FJ, Chen J, Liang P, Narciso C, Levis MK, Arredondo-Walsh N, Chen DZ, Zartman JJ. Decoding Calcium Signaling Dynamics during Drosophila Wing Disc Development. Biophys J 2019; 116:725-740. [PMID: 30704858 PMCID: PMC6382932 DOI: 10.1016/j.bpj.2019.01.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/04/2018] [Accepted: 01/04/2019] [Indexed: 01/07/2023] Open
Abstract
The robust specification of organ development depends on coordinated cell-cell communication. This process requires signal integration among multiple pathways, relying on second messengers such as calcium ions. Calcium signaling encodes a significant portion of the cellular state by regulating transcription factors, enzymes, and cytoskeletal proteins. However, the relationships between the inputs specifying cell and organ development, calcium signaling dynamics, and final organ morphology are poorly understood. Here, we have designed a quantitative image-analysis pipeline for decoding organ-level calcium signaling. With this pipeline, we extracted spatiotemporal features of calcium signaling dynamics during the development of the Drosophila larval wing disc, a genetic model for organogenesis. We identified specific classes of wing phenotypes that resulted from calcium signaling pathway perturbations, including defects in gross morphology, vein differentiation, and overall size. We found four qualitative classes of calcium signaling activity. These classes can be ordered based on agonist stimulation strength Gαq-mediated signaling. In vivo calcium signaling dynamics depend on both receptor tyrosine kinase/phospholipase C γ and G protein-coupled receptor/phospholipase C β activities. We found that spatially patterned calcium dynamics correlate with known differential growth rates between anterior and posterior compartments. Integrated calcium signaling activity decreases with increasing tissue size, and it responds to morphogenetic perturbations that impact organ growth. Together, these findings define how calcium signaling dynamics integrate upstream inputs to mediate multiple response outputs in developing epithelial organs.
Collapse
Affiliation(s)
- Pavel A Brodskiy
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Qinfeng Wu
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Dharsan K Soundarrajan
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Francisco J Huizar
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Jianxu Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Peixian Liang
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Cody Narciso
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Megan K Levis
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana
| | | | - Danny Z Chen
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Jeremiah J Zartman
- Department of Chemical and Biomolecular Engineering, University of Notre Dame, Notre Dame, Indiana.
| |
Collapse
|
21
|
Mathews J, Levin M. The body electric 2.0: recent advances in developmental bioelectricity for regenerative and synthetic bioengineering. Curr Opin Biotechnol 2018; 52:134-144. [PMID: 29684787 PMCID: PMC10464502 DOI: 10.1016/j.copbio.2018.03.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/23/2018] [Indexed: 12/18/2022]
Abstract
Breakthroughs in biomedicine and synthetic bioengineering require predictive, rational control over anatomical structure and function. Recent successes in manipulating cellular and molecular hardware have not been matched by progress in understanding the patterning software implemented during embryogenesis and regeneration. A fundamental capability gap is driving desired changes in growth and form to address birth defects and traumatic injury. Here we review new tools, results, and conceptual advances in an exciting emerging field: endogenous non-neural bioelectric signaling, which enables cellular collectives to make global decisions and implement large-scale pattern homeostasis. Spatially distributed electric circuits regulate gene expression, organ morphogenesis, and body-wide axial patterning. Developmental bioelectricity facilitates the interface to organ-level modular control points that direct patterning in vivo. Cracking the bioelectric code will enable transformative progress in bioengineering and regenerative medicine.
Collapse
Affiliation(s)
- Juanita Mathews
- Biology Department, and Allen Discovery Center at Tufts University, Medford, MA 02155, United States
| | - Michael Levin
- Biology Department, and Allen Discovery Center at Tufts University, Medford, MA 02155, United States.
| |
Collapse
|