1
|
McGehee J, Stathopoulos A. Mechanisms for controlling Dorsal nuclear levels. Front Cell Dev Biol 2024; 12:1436369. [PMID: 39161589 PMCID: PMC11330768 DOI: 10.3389/fcell.2024.1436369] [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: 05/22/2024] [Accepted: 07/18/2024] [Indexed: 08/21/2024] Open
Abstract
Formation of the Dorsal nuclear-cytoplasmic gradient is important for the proper establishment of gene expression patterns along the dorsal-ventral (DV) axis during embryogenesis in Drosophila melanogaster. Correct patterning of the DV axis leads to formation of the presumptive mesoderm, neurogenic ectoderm, dorsal ectoderm, and amnioserosa, which are tissues necessary for embryo viability. While Toll signaling is necessary for Dorsal gradient formation, a gradient still forms in the absence of Toll, suggesting there are additional mechanisms required to achieve correct nuclear Dorsal levels. Potential mechanisms include post-translational modification, shuttling, and nuclear spacing. Post-translational modification could affect import and export rates either directly through modification of a nuclear localization sequence or nuclear export sequence, or indirectly by affecting interactions with binding partners that alter import and export rates. Shuttling, which refers to the facilitated diffusion of Dorsal through its interaction with its cytoplasmic inhibitor Cactus, could regulate nuclear levels by delivering more Dorsal ventrally. Finally, nuclear spacing could result in higher nuclear levels by leaving fewer nuclei in the ventral domain to uptake Dorsal. This review details how each of these mechanisms may help establish Dorsal nuclear levels in the early fly embryo, which serves as a paradigm for understanding how the dynamics of graded inputs can influence patterning and target gene expression. Furthermore, careful analysis of nuclear Dorsal levels is likely to provide general insights as recent studies have suggested that the regulation of nuclear import affects the timing of gene expression at the maternal-to-zygotic transition.
Collapse
Affiliation(s)
| | - Angelike Stathopoulos
- California Institute of Technology, Division of Biology and Biological Engineering, Pasadena, CA, United States
| |
Collapse
|
2
|
Liu S, Ge D, Long Z, Chi C, Lv Z, Liu H. Molecular features of interleukin-1 receptor-associated kinase-b and a in Mytilus coruscus, regulating their function by infection of aquatic pathogens and the expression of their serine/threonine protein kinase functional domains. FISH & SHELLFISH IMMUNOLOGY 2020; 102:469-479. [PMID: 32389741 DOI: 10.1016/j.fsi.2020.05.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 05/01/2020] [Accepted: 05/03/2020] [Indexed: 06/11/2023]
Abstract
Interleukin-1 receptor-associated kinases (IRAKs) play important roles in the innate immune system of TLR (Toll-like receptor) signaling pathway. In this paper, interleukin-1 receptor-associated kinase-b (designated as McIRAK-b) and interleukin-1 receptor-associated kinase-a (named as McIRAK-a) were obtained based on the transcriptome data, the full length of McIRAK-b was 1815 bp and McIRAK-a was 3168bp, encoding 532 and 978 amino acids, respectively. BLASTp analysis and phylogenetic relationship strongly suggested that the deduced amino acid sequence of McIRAK-b had high homology with IRAK-4 and McIRAK-a was similar to IRAK-1 of other mollusks, especially at their function domains. The expressions of McIRAK-b and McIRAK-a were detected in six tissues including adductor muscle, hemocyte, gills, gonad and hepatopancreas, and the highest expressions appeared both in gills. The expressions of McIRAK-b and McIRAK-a in gills were observed with time-dependent manners after bacterial infections. After being challenged with Vibrio alginolyticus, McIRAK-b expressed significantly and got the peak at 8 h (9.47 times compared with the control group), but the peak appeared at 4 h by being infected with Vibrio parahaemolyticus (12.02 times compared with the control group). The highest point of McIRAK-a mRNA appeared at 12 h (5.16 times) after being challenged with V.alginolyticus and 8 h (4.21 times) for V.parahaemolyticus challenge. The results suggested that IRAK-b and IRAK-a might be important in immune signaling pathway of mussels. The kinase functional domain sequences (S_TKc) of McIRAK-b and McIRAK-a expressed in BL21(DE3) and purified by Ni-NAT Superflow resin conforming to the expected molecular weight with many active sites for their conferring protein-protein interaction functions. This study may provide some further understandings of the regulatory mechanisms in the bivalve innate immune system for IRAKs family.
Collapse
Affiliation(s)
- Sijia Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Delong Ge
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Zaihao Long
- Ningbo International Travel Health Care Center, Ningbo, 315012, PR China
| | - Changfeng Chi
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Zhenming Lv
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China
| | - Huihui Liu
- National and Provincial Joint Laboratory of Exploration and Utilization of Marine Aquatic Genetic Resources, National Engineering Research Center of Marine Facilities Aquaculture, Zhejiang Ocean University, Zhoushan, 316022, PR China.
| |
Collapse
|
3
|
Ho CH, Treisman JE. Specific Isoforms of the Guanine-Nucleotide Exchange Factor dPix Couple Neuromuscular Synapse Growth to Muscle Growth. Dev Cell 2020; 54:117-131.e5. [PMID: 32516570 DOI: 10.1016/j.devcel.2020.05.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/09/2020] [Accepted: 05/13/2020] [Indexed: 12/28/2022]
Abstract
Developmental growth requires coordination between the growth rates of individual tissues and organs. Here, we examine how Drosophila neuromuscular synapses grow to match the size of their target muscles. We show that changes in muscle growth driven by autonomous modulation of insulin receptor signaling produce corresponding changes in synapse size, with each muscle affecting only its presynaptic motor neuron branches. This scaling growth is mechanistically distinct from synaptic plasticity driven by neuronal activity and requires increased postsynaptic differentiation induced by insulin receptor signaling in muscle. We identify the guanine-nucleotide exchange factor dPix as an effector of insulin receptor signaling. Alternatively spliced dPix isoforms that contain a specific exon are necessary and sufficient for postsynaptic differentiation and scaling growth, and their mRNA levels are regulated by insulin receptor signaling. These findings define a mechanism by which the same signaling pathway promotes both autonomous muscle growth and non-autonomous synapse growth.
Collapse
Affiliation(s)
- Cheuk Hei Ho
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine at the Skirball Institute and Department of Cell Biology, NYU School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
4
|
Schloop AE, Bandodkar PU, Reeves GT. Formation, interpretation, and regulation of the Drosophila Dorsal/NF-κB gradient. Curr Top Dev Biol 2019; 137:143-191. [PMID: 32143742 DOI: 10.1016/bs.ctdb.2019.11.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The morphogen gradient of the transcription factor Dorsal in the early Drosophila embryo has become one of the most widely studied tissue patterning systems. Dorsal is a Drosophila homolog of mammalian NF-κB and patterns the dorsal-ventral axis of the blastoderm embryo into several tissue types by spatially regulating upwards of 100 zygotic genes. Recent studies using fluorescence microscopy and live imaging have quantified the Dorsal gradient and its target genes, which has paved the way for mechanistic modeling of the gradient. In this review, we describe the mechanisms behind the initiation of the Dorsal gradient and its regulation of target genes. The main focus of the review is a discussion of quantitative and computational studies of the Dl gradient system, including regulation of the Dl gradient. We conclude with a discussion of potential future directions.
Collapse
Affiliation(s)
- Allison E Schloop
- Genetics Program, North Carolina State University, Raleigh, NC, United States
| | - Prasad U Bandodkar
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, United States
| | - Gregory T Reeves
- Genetics Program, North Carolina State University, Raleigh, NC, United States; Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC, United States.
| |
Collapse
|
5
|
Tang X, Huang B, Zhang L, Li L, Zhang G. Molecular characterization of Pacific oyster (Crassostrea gigas) IRAK4 gene and its role in MyD88-dependent pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 72:21-29. [PMID: 28223161 DOI: 10.1016/j.dci.2017.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/09/2017] [Accepted: 02/09/2017] [Indexed: 06/06/2023]
Abstract
Interleukin-1 receptor-associated kinases (IRAKs) play important roles in MyD88-dependent TLR signaling, the crucial innate immune pathway in molluscs. In this study, we examined the full-length IRAK4 genetic sequence in the Pacific oyster (Crassostrea gigas) by molecular cloning. Phylogenetic analysis revealed that CgIRAK4 is most closely related to Mytilus edulis, and forms a clade with other molluscs. CgIRAK4 transcripts are widely expressed in all tissues, with the highest expression observed in the hemocytes and gill. Moreover, CgIRAK4 is significantly upregulated after Oyster herpesvirus-1 microvariant (OsHV-1 μvar), Vibrio alginolyticus, and poly I:C challenge. Yeast two-hybrid and co-immunoprecipitation assays reveal that the CgIRAK4 death domain is necessary to mediate interaction between CgIRAK4 and two CgMyD88 isoforms. In addition, CgIRAK4 overexpression cannot induce NF-κB transcriptional activity, but blocks that induced by CgMyD88 in HEK293T cells. These findings elucidate the mechanisms of MyD88-dependent TLR signaling in molluscs, and the differences in IRAK-mediated pathway activation between invertebrates and vertebrates.
Collapse
Affiliation(s)
- Xueying Tang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong 266071, China; University of Chinese Academy of Sciences, Beijing 100039, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, Shandong 266071, China
| | - Baoyu Huang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, Shandong 266071, China
| | - Linlin Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, Shandong 266071, China
| | - Li Li
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Fisheries and Aquaculture, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, Shandong 266071, China
| | - Guofan Zhang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, Shandong 266071, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong 266071, China; National & Local Joint Engineering Laboratory of Ecological Mariculture, Qingdao, Shandong 266071, China.
| |
Collapse
|
6
|
Zhou B, Lindsay SA, Wasserman SA. Alternative NF-κB Isoforms in the Drosophila Neuromuscular Junction and Brain. PLoS One 2015; 10:e0132793. [PMID: 26167685 PMCID: PMC4500392 DOI: 10.1371/journal.pone.0132793] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/19/2015] [Indexed: 11/19/2022] Open
Abstract
The Drosophila NF-κB protein Dorsal is expressed at the larval neuromuscular junction, where its expression appears unrelated to known Dorsal functions in embryonic patterning and innate immunity. Using confocal microscopy with domain-specific antisera, we demonstrate that larval muscle expresses only the B isoform of Dorsal, which arises by intron retention. We find that Dorsal B interacts with and stabilizes Cactus at the neuromuscular junction, but exhibits Cactus independent localization and an absence of detectable nuclear translocation. We further find that the Dorsal-related immune factor Dif encodes a B isoform, reflecting a conservation of B domains across a range of insect NF-κB proteins. Carrying out mutagenesis of the Dif locus via a site-specific recombineering approach, we demonstrate that Dif B is the major, if not sole, Dif isoform in the mushroom bodies of the larval brain. The Dorsal and Dif B isoforms thus share a specific association with nervous system tissues as well as an alternative protein structure.
Collapse
Affiliation(s)
- Bo Zhou
- Section of Cell & Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Scott A. Lindsay
- Section of Cell & Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Steven A. Wasserman
- Section of Cell & Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
7
|
Li YX, Dijkers PF. Specific Calcineurin Isoforms Are Involved in Drosophila Toll Immune Signaling. THE JOURNAL OF IMMUNOLOGY 2014; 194:168-76. [DOI: 10.4049/jimmunol.1401080] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
8
|
Stein DS, Stevens LM. Maternal control of the Drosophila dorsal-ventral body axis. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:301-30. [PMID: 25124754 DOI: 10.1002/wdev.138] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Revised: 03/24/2014] [Accepted: 04/07/2014] [Indexed: 12/14/2022]
Abstract
UNLABELLED The pathway that generates the dorsal-ventral (DV) axis of the Drosophila embryo has been the subject of intense investigation over the previous three decades. The initial asymmetric signal originates during oogenesis by the movement of the oocyte nucleus to an anterior corner of the oocyte, which establishes DV polarity within the follicle through signaling between Gurken, the Drosophila Transforming Growth Factor (TGF)-α homologue secreted from the oocyte, and the Drosophila Epidermal Growth Factor Receptor (EGFR) that is expressed by the follicular epithelium cells that envelop the oocyte. Follicle cells that are not exposed to Gurken follow a ventral fate and express Pipe, a sulfotransferase that enzymatically modifies components of the inner vitelline membrane layer of the eggshell, thereby transferring DV spatial information from the follicle to the egg. These ventrally sulfated eggshell proteins comprise a localized cue that directs the ventrally restricted formation of the active Spätzle ligand within the perivitelline space between the eggshell and the embryonic membrane. Spätzle activates Toll, a transmembrane receptor in the embryonic membrane. Transmission of the Toll signal into the embryo leads to the formation of a ventral-to-dorsal gradient of the transcription factor Dorsal within the nuclei of the syncytial blastoderm stage embryo. Dorsal controls the spatially specific expression of a large constellation of zygotic target genes, the Dorsal gene regulatory network, along the embryonic DV circumference. This article reviews classic studies and integrates them with the details of more recent work that has advanced our understanding of the complex pathway that establishes Drosophila embryo DV polarity. For further resources related to this article, please visit the WIREs website. CONFLICT OF INTEREST The authors have declared no conflicts of interest for this article.
Collapse
Affiliation(s)
- David S Stein
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | | |
Collapse
|
9
|
The IRAK homolog Pelle is the functional counterpart of IκB kinase in the Drosophila Toll pathway. PLoS One 2013; 8:e75150. [PMID: 24086459 PMCID: PMC3781037 DOI: 10.1371/journal.pone.0075150] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 08/09/2013] [Indexed: 12/30/2022] Open
Abstract
Toll receptors transduce signals that activate Rel-family transcription factors, such as NF-κB, by directing proteolytic degradation of inhibitor proteins. In mammals, the IκB Kinase (IKK) phosphorylates the inhibitor IκBα. A βTrCP protein binds to phosphorylated IκBα, triggering ubiquitination and proteasome mediated degradation. In Drosophila, Toll signaling directs Cactus degradation via a sequence motif that is highly similar to that in IκBα, but without involvement of IKK. Here we show that Pelle, the homolog of a mammalian regulator of IKK, acts as a Cactus kinase. We further find that the fly βTrCP protein Slimb is required in cultured cells to mediate Cactus degradation. These findings enable us for the first time to trace an uninterrupted pathway from the cell surface to the nucleus for Drosophila Toll signaling.
Collapse
|
10
|
Fontenele M, Lim B, Oliveira D, Buffolo M, Perlman DH, Schupbach T, Araujo H. Calpain A modulates Toll responses by limited Cactus/IκB proteolysis. Mol Biol Cell 2013; 24:2966-80. [PMID: 23864715 PMCID: PMC3771957 DOI: 10.1091/mbc.e13-02-0113] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Calcium-dependent cysteine proteases of the calpain family are modulatory proteases that cleave their substrates in a limited manner. Among their substrates, calpains target vertebrate and invertebrate IκB proteins. Because proteolysis by calpains potentially generates novel protein functions, it is important to understand how this affects NFκB activity. We investigate the action of Calpain A (CalpA) on the Drosophila melanogaster IκB homologue Cactus in vivo. CalpA alters the absolute amounts of Cactus protein. Our data indicate, however, that CalpA uses additional mechanisms to regulate NFκB function. We provide evidence that CalpA interacts physically with Cactus, recognizing a Cactus pool that is not bound to Dorsal, a fly NFκB/Rel homologue. We show that proteolytic cleavage by CalpA generates Cactus fragments lacking an N-terminal region required for Toll responsiveness. These fragments are generated in vivo and display properties distinct from those of full-length Cactus. We propose that CalpA targets free Cactus, which is incorporated into and modulates Toll-responsive complexes in the embryo and immune system.
Collapse
Affiliation(s)
- Marcio Fontenele
- Institute for Biomedical Sciences, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Brazil Chemistry Institute, Federal University of Rio de Janeiro, CEP 21941-902 Rio de Janeiro, Brazil Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Rio de Janeiro, Brazil Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544 Princeton Collaborative Proteomics and Mass Spectrometry Center, Princeton University, Princeton, NJ 08544 Molecular Biology Department, Princeton University, Princeton, NJ 08544 Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
The vast majority of research on nuclear factor κB (NF-κB) signaling in the past 25 years has focused on its roles in normal and disease-related processes in vertebrates, especially mice and humans. Recent genome and transcriptome sequencing efforts have shown that homologs of NF-κB transcription factors, inhibitor of NF-κB (IκB) proteins, and IκB kinases are present in a variety of invertebrates, including several in phyla simpler than Arthropoda, the phylum containing insects such Drosophila. Moreover, many invertebrates also contain genes encoding homologs of upstream signaling proteins in the Toll-like receptor signaling pathway, which is well-known for its downstream activation of NF-κB for innate immunity. This review describes what we now know or can infer and speculate about the evolution of the core elements of NF-κB signaling as well as the biological processes controlled by NF-κB in invertebrates. Further research on NF-κB in invertebrates is likely to uncover information about the evolutionary origins of this key human signaling pathway and may have relevance to our management of the responses of ecologically and economically important organisms to environmental and adaptive pressures.
Collapse
Affiliation(s)
- Thomas D Gilmore
- Department of Biology, Boston University, Boston, MA 02215, USA.
| | | |
Collapse
|
12
|
Schiffmann Y. Maternal-effect genes as the recording genes of Turing-Child patterns: Sequential compartmentalization in Drosophila. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2012; 109:16-32. [DOI: 10.1016/j.pbiomolbio.2012.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 04/18/2012] [Indexed: 10/28/2022]
|
13
|
Chen J, Lippincott-Schwartz J, Liu J. Intracellular spatial localization regulated by the microtubule network. PLoS One 2012; 7:e34919. [PMID: 22532834 PMCID: PMC3330817 DOI: 10.1371/journal.pone.0034919] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 03/06/2012] [Indexed: 11/17/2022] Open
Abstract
The commonly recognized mechanisms for spatial regulation inside the cell are membrane-bounded compartmentalization and biochemical association with subcellular organelles. We use computational modeling to investigate another spatial regulation mechanism mediated by the microtubule network in the cell. Our results demonstrate that the mitotic spindle can impose strong sequestration and concentration effects on molecules with binding affinity for microtubules, especially dynein-directed cargoes. The model can recapitulate the essence of three experimental observations on distinct microtubule network morphologies: the sequestration of germ plasm components by the mitotic spindles in the Drosophila syncytial embryo, the asymmetric cell division initiated by the time delay in centrosome maturation in the Drosophila neuroblast, and the diffusional block between neighboring energids in the Drosophila syncytial embryo. Our model thus suggests that the cell cycle-dependent changes in the microtubule network are critical for achieving different spatial regulation effects. The microtubule network provides a spatially extensive docking platform for molecules and gives rise to a "structured cytoplasm", in contrast to a free and fluid environment.
Collapse
Affiliation(s)
- Jing Chen
- National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | | | | |
Collapse
|
14
|
Drosophila embryo syncytial blastoderm cellular architecture and morphogen gradient dynamics: Is there a correlation? ACTA ACUST UNITED AC 2012. [DOI: 10.1007/s11515-011-1160-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
15
|
Ganesan S, Aggarwal K, Paquette N, Silverman N. NF-κB/Rel proteins and the humoral immune responses of Drosophila melanogaster. Curr Top Microbiol Immunol 2011; 349:25-60. [PMID: 20852987 PMCID: PMC3083852 DOI: 10.1007/82_2010_107] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Nuclear Factor-κB (NF-κB)/Rel transcription factors form an integral part of innate immune defenses and are conserved throughout the animal kingdom. Studying the function, mechanism of activation and regulation of these factors is crucial for understanding host responses to microbial infections. The fruit fly Drosophila melanogaster has proved to be a valuable model system to study these evolutionarily conserved NF-κB mediated immune responses. Drosophila combats pathogens through humoral and cellular immune responses. These humoral responses are well characterized and are marked by the robust production of a battery of anti-microbial peptides. Two NF-κB signaling pathways, the Toll and the IMD pathways, are responsible for the induction of these antimicrobial peptides. Signal transduction in these pathways is strikingly similar to that in mammalian TLR pathways. In this chapter, we discuss in detail the molecular mechanisms of microbial recognition, signal transduction and NF-κB regulation, in both the Toll and the IMD pathways. Similarities and differences relative to their mammalian counterparts are discussed, and recent advances in our understanding of the intricate regulatory networks in these NF-κB signaling pathways are also highlighted.
Collapse
Affiliation(s)
- Sandhya Ganesan
- Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | |
Collapse
|
16
|
Fontenele M, Carneiro K, Agrellos R, Oliveira D, Oliveira-Silva A, Vieira V, Negreiros E, Machado E, Araujo H. The Ca2+-dependent protease Calpain A regulates Cactus/I kappaB levels during Drosophila development in response to maternal Dpp signals. Mech Dev 2009; 126:737-51. [PMID: 19442719 DOI: 10.1016/j.mod.2009.04.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2009] [Revised: 04/24/2009] [Accepted: 04/27/2009] [Indexed: 11/15/2022]
Abstract
Regulation of NF kappaB activity is central to many processes during development and disease. Activation of NF kappaB family members depends on degradation of inhibitory I kappaB proteins. In Drosophila, a nuclear gradient of the NF kappaB/c-rel protein Dorsal subdivides the embryonic dorsal-ventral axis, defining the extent and location of mesodermal and ectodermal territories. Activation of the Toll pathway directs Dorsal nuclear translocation by inducing proteosomal degradation of the I kappaB homologue Cactus. Another mechanism that impacts on Dorsal activation involves the Toll-independent pathway, which regulates constitutive Cactus degradation. We have shown that the BMP protein Decapentaplegic (Dpp) inhibits Cactus degradation independent of Toll. Here we report on a novel element of this pathway: the calcium-dependent protease Calpain A. Calpain A knockdowns increase Cactus levels, shifting the Dorsal gradient and dorsal-ventral patterning. As shown for mammalian I kappaB, this effect requires PEST sequences in the Cactus C-terminus, implying a conserved role for calpains. Alteration of Calpain A or dpp results in similar effects on Dorsal target genes. Epistatic analysis confirms Calpain A activity is regulated by Dpp, indicating that Dpp signals increase Cactus levels through Calpain A inhibition, thereby interfering with Dorsal activation. This mechanism may allow coordination of Toll, BMP and Ca(2+) signals, conferring precision to Dorsal-target expression domains.
Collapse
Affiliation(s)
- M Fontenele
- Institute for Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
DeLotto R, DeLotto Y, Steward R, Lippincott-Schwartz J. Nucleocytoplasmic shuttling mediates the dynamic maintenance of nuclear Dorsal levels during Drosophila embryogenesis. Development 2007; 134:4233-41. [PMID: 17978003 DOI: 10.1242/dev.010934] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
In Drosophila, the NF-kappaB/REL family transcription factor, Dorsal, redistributes from the cytoplasm to nuclei, forming a concentration gradient across the dorsoventral axis of the embryo. Using live imaging techniques in conjunction with embryos expressing a chimeric Dorsal-GFP, we demonstrate that the redistribution of Dorsal from cytoplasm to nucleus is an extremely dynamic process. Nuclear Dorsal concentration changes continuously over time in all nuclei during interphase. While Dorsal appears to be nuclearly localized primarily in ventral nuclei, it is actively shuttling into and out of all nuclei, including nuclei on the dorsal side. Nuclear export is blocked by leptomycin B, a potent inhibitor of Exportin 1 (CRM1)-mediated nuclear export. We have developed a novel in vivo assay revealing the presence of a functional leucine-rich nuclear export signal within the carboxyterminal 44 amino acids of Dorsal. We also find that diffusion of Dorsal is partially constrained to cytoplasmic islands surrounding individual syncitial nuclei. A model is proposed in which the generation and maintenance of the Dorsal gradient is a consequence of an active process involving both restricted long-range diffusion and the balancing of nuclear import with nuclear export.
Collapse
Affiliation(s)
- Robert DeLotto
- Department of Molecular Biology, University of Copenhagen, Ole Maaløes Vej 5, DK-2200 Copenhagen N, Denmark.
| | | | | | | |
Collapse
|
18
|
Heckscher ES, Fetter RD, Marek KW, Albin SD, Davis GW. NF-kappaB, IkappaB, and IRAK control glutamate receptor density at the Drosophila NMJ. Neuron 2007; 55:859-73. [PMID: 17880891 PMCID: PMC2701504 DOI: 10.1016/j.neuron.2007.08.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Revised: 06/04/2007] [Accepted: 08/07/2007] [Indexed: 10/22/2022]
Abstract
NF-kappaB signaling has been implicated in neurodegenerative disease, epilepsy, and neuronal plasticity. However, the cellular and molecular activity of NF-kappaB signaling within the nervous system remains to be clearly defined. Here, we show that the NF-kappaB and IkappaB homologs Dorsal and Cactus surround postsynaptic glutamate receptor (GluR) clusters at the Drosophila NMJ. We then show that mutations in dorsal, cactus, and IRAK/pelle kinase specifically impair GluR levels, assayed immunohistochemically and electrophysiologically, without affecting NMJ growth, the size of the postsynaptic density, or homeostatic plasticity. Additional genetic experiments support the conclusion that cactus functions in concert with, rather than in opposition to, dorsal and pelle in this process. Finally, we provide evidence that Dorsal and Cactus act posttranscriptionally, outside the nucleus, to control GluR density. Based upon our data we speculate that Dorsal, Cactus, and Pelle could function together, locally at the postsynaptic density, to specify GluR levels.
Collapse
Affiliation(s)
- Elizabeth S Heckscher
- Department of Biochemistry and Biophysics, Programs in Neuroscience and Cell Biology, University of California, San Francisco, 1550 4th Street, San Francisco, CA 94158-2822, USA
| | | | | | | | | |
Collapse
|
19
|
Beramendi A, Peron S, Casanova G, Reggiani C, Cantera R. Neuromuscular junction in abdominal muscles ofDrosophila melanogasterduring adulthood and aging. J Comp Neurol 2007; 501:498-508. [PMID: 17278125 DOI: 10.1002/cne.21253] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The neuromuscular junction (NMJ) of Drosophila melanogaster has been established as a productive model for the study of synaptogenesis, synaptic plasticity, vesicle recycling, and other synaptic functions in embryos and larvae. It also has potential for the study of long-term plasticity during adult life and degenerative processes associated with aging. Here we provide a detailed description of the morphology and ultrastructure of the NMJ on abdominal dorsal longitudinal muscles throughout adult life from eclosion to senescence. In contrast to the case in the larva, the predominant type of terminals in these muscles in the adult fly consists of only two or three branches with tightly packed synaptic boutons. We observed qualitative and quantitative changes as mean bouton size increased gradually during adulthood, and the largest boutons were present in the old fly. The length of nerve branches first increased and thereafter decreased gradually during most of adult life. Branch diameter also decreased progressively, but branch number did not change. The subsynaptic reticulum became progressively thinner, and "naked" boutons were found in old flies. Ultrastructural traits gave indications of an age-associated increment in autophagy, larger synaptic vesicles, and impaired endocytosis. We propose that NMJ aging in the fly correlates with impaired endocytosis and membrane dynamics. This view finds a functional correlate in flies carrying a temperature-sensitive mutation in shibire that reversible blocks endocytosis; age significantly reduces the time required for complete paralysis and increases the time of recovery, thus confirming the age-dependent alteration in vesicle dynamics.
Collapse
Affiliation(s)
- Ana Beramendi
- Department of Zoology, Stockholm University, S-106 91 Stockholm, Sweden 35131
| | | | | | | | | |
Collapse
|
20
|
Harari-Steinberg O, Cantera R, Denti S, Bianchi E, Oron E, Segal D, Chamovitz DA. COP9 signalosome subunit 5 (CSN5/Jab1) regulates the development of the Drosophila immune system: effects on Cactus, Dorsal and hematopoiesis. Genes Cells 2007; 12:183-95. [PMID: 17295838 DOI: 10.1111/j.1365-2443.2007.01049.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The COP9 signalosome is a multifunctional regulator essential for Drosophila development. A loss-of-function mutant in Drosophila COP9 signalosome subunit 5 (CSN5) develops melanotic bodies, a phenotype common to mutants in immune signaling. csn5(null) larvae accumulated high levels of Cactus that co-localizes with Dorsal to the nucleus. However, Dorsal-dependent transcriptional activity remained repressed in the absence of an inducing signal, despite its nuclear localization. Dorsal activity in mutant larvae and NFkappaB activity in CSN5 down-regulated mammalian cells can be induced following activation of the Toll/IL-1 pathway. csn5(null) larvae contained more hemocytes than wild-type (wt) larvae. A large portion of these cells have differentiated to lamellocytes (LM), a hemocyte cell type rarely seen in normal larvae. The results presented here indicate that CSN5 is a negative regulator of Dorsal subcellular localization, and of hemocyte proliferation and differentiation. These results further indicate that nuclear localization of Dorsal can be uncoupled from its activation. Surprisingly, CSN5 is not necessary for immune-induced degradation of Cactus.
Collapse
|
21
|
Abstract
The nuclear factor kappa B (NF-kappaB) pathways in Drosophila are multi-component pathways, as in vertebrates, that regulate the expression of many genes responsible for the formation of dorsal-ventral polarity in the early embryo, the innate immune response to infection with Gram- negative and positive bacteria and fungi, the cellular immune response and hematopoiesis. Overactivation of the fly pathway can result in developmental defects, overproliferation of hemocytes and the formation of melanotic tumors or nodules. The extracellular events leading to the maturation of the ligand for initiation of the Drosophila NF-kappaB pathway is not conserved between flies and vertebrates, but the Toll receptor and downstream events are remarkably similar. NF-kappaB proteins have been identified in mollusks, and arthropods such as horseshoe crabs and beetles, indicating that this pathway has been established more than 500 million years ago. The fly NF-kappaB pathways are less complex than those in vertebrates, with the involvement of fewer proteins, but they are, nonetheless, just as important as their vertebrate counterparts for the life of the fly.
Collapse
Affiliation(s)
- S Minakhina
- Waksman Institute, Department of Molecular Biology and Biochemistry and the Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ 08854, USA
| | | |
Collapse
|
22
|
Xylourgidis N, Roth P, Sabri N, Tsarouhas V, Samakovlis C. The nucleoporin Nup214 sequesters CRM1 at the nuclear rim and modulates NFkappaB activation in Drosophila. J Cell Sci 2006; 119:4409-19. [PMID: 17032737 DOI: 10.1242/jcs.03201] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
CRM1-mediated protein export is an important determinant of the nuclear accumulation of many gene regulators. Here, we show that the NFkappaB transcription factor Dorsal is a substrate of CRM1 and requires the nucleoporin Nup214 for its nuclear translocation upon signaling. Nup214 bound to CRM1 directly and anchored it to the nuclear envelope. In nup214 mutants CRM1 accumulated in the nucleus and NES-protein export was enhanced. Nup214 formed complexes with Nup88 and CRM1 in vivo and Nup214 protected Nup88 from degradation at the nuclear rim. In turn, Nup88 was sufficient for targeting the complex to the nuclear pores. Overexpression experiments indicated that Nup214 alone attracts a fraction of CRM1 to the nuclear envelope but does not interfere with NES-GFP export. By contrast, overexpression of the Nup214-Nup88 complex trapped CRM1 and Dorsal to cytoplasmic foci and inhibited protein export and immune response activation. We hypothesize that variation in levels of the Nup214-Nup88 complex at the pore changes the amount of NPC-bound CRM1 and influences the relative strength and duration of NFkappaB signaling responses.
Collapse
Affiliation(s)
- Nikos Xylourgidis
- Department of Developmental Biology, Wenner-Gren Institute, Stockholm University, S-10691, Stockholm, Sweden
| | | | | | | | | |
Collapse
|
23
|
Gordon MD, Dionne MS, Schneider DS, Nusse R. WntD is a feedback inhibitor of Dorsal/NF-kappaB in Drosophila development and immunity. Nature 2005; 437:746-9. [PMID: 16107793 PMCID: PMC1256032 DOI: 10.1038/nature04073] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2005] [Accepted: 07/20/2005] [Indexed: 11/10/2022]
Abstract
Regulating the nuclear factor-kappaB (NF-kappaB) family of transcription factors is of critical importance to animals, with consequences of misregulation that include cancer, chronic inflammatory diseases and developmental defects. Studies in Drosophila melanogaster have proved fruitful in determining the signals used to control NF-kappaB proteins, beginning with the discovery that the Toll/NF-kappaB pathway, in addition to patterning the dorsal-ventral axis of the fly embryo, defines a major component of the innate immune response in both Drosophila and mammals. Here, we characterize the Drosophila wntD (Wnt inhibitor of Dorsal) gene. We show that WntD acts as a feedback inhibitor of the NF-kappaB homologue Dorsal during both embryonic patterning and the innate immune response to infection. wntD expression is under the control of Toll/Dorsal signalling, and increased levels of WntD block Dorsal nuclear accumulation, even in the absence of the IkappaB homologue Cactus. The WntD signal is independent of the common Wnt signalling component Armadillo (beta-catenin). By engineering a gene knockout, we show that wntD loss-of-function mutants have immune defects and exhibit increased levels of Toll/Dorsal signalling. Furthermore, the wntD mutant phenotype is suppressed by loss of zygotic dorsal. These results describe the first secreted feedback antagonist of Toll signalling, and demonstrate a novel Wnt activity in the fly.
Collapse
Affiliation(s)
- Michael D Gordon
- Department of Developmental Biology, Howard Hughes Medical Institute, Beckman Center and
| | | | | | | |
Collapse
|
24
|
Beramendi A, Peron S, Megighian A, Reggiani C, Cantera R. The inhibitorκB-ortholog Cactus is necessary for normal neuromuscular function in Drosophila melanogaster. Neuroscience 2005; 134:397-406. [PMID: 15975723 DOI: 10.1016/j.neuroscience.2005.04.046] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2005] [Revised: 04/22/2005] [Accepted: 04/23/2005] [Indexed: 10/25/2022]
Abstract
The Drosophila inhibitor-kappaB ortholog Cactus acts as an inhibitor of the Rel-transcription factors Dorsal and Dif. In blastoderm cells and immune competent cells, Cactus inhibits Dorsal and Dif by preventing their nuclear localization. Cactus, Dorsal and Dif are also expressed in somatic muscles, where Cactus and Dorsal, but not Dif, are enriched at the neuromuscular junction. Mutations in dorsal cause neuromuscular defects and mislocalization of Cactus. Here, we investigated whether mutations in cactus affect the neuromuscular system and subcellular localization of Dorsal and Dif. Using locomotion assays, as well as physiological and immunochemical methods, we found that wild type Cactus is necessary for the normal function of the larval neuromuscular system. The phenotype comprises i) altered bouton numbers and impaired neurotransmitter release in the neuromuscular junctions in the abdominal segments, ii) muscular weakness and iii) poor locomotion performance, probably reflecting a general neuromuscular impairment. Interestingly, in cactus mutants the subcellular localization of Dorsal and Dif in muscle is not affected, whereas cactus protein is not detected in the nucleus. This suggests, together with the similarities between the phenotypes induced by cactus and dorsal mutations, that in larval muscles the function of Cactus might be cooperation to the transcriptional activity of Rel proteins more than their cytoplasmic retention. The similarities with inhibitor-kappaB/nuclear factor kappaB interactions and muscle pathology in mammals point to Drosophila as a suitable experimental system to clarify the complex interactions of these proteins in muscle postembryonic development and activity.
Collapse
Affiliation(s)
- A Beramendi
- Department of Zoology, Stockholm University, S-106 91 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
25
|
Lopes ESS, Araujo HM. The maternal JAK/STAT pathway of Drosophila regulates embryonic dorsal-ventral patterning. Braz J Med Biol Res 2004; 37:1811-8. [PMID: 15558187 DOI: 10.1590/s0100-879x2004001200006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Activation of NFkappaB plays a pivotal role in many cellular processes such as inflammation, proliferation and apoptosis. In Drosophila, nuclear translocation of the NFkappaB-related transcription factor Dorsal is spatially regulated in order to subdivide the embryo into three primary dorsal-ventral (DV) domains: the ventral presumptive mesoderm, the lateral neuroectoderm and the dorsal ectoderm. Ventral activation of the Toll receptor induces degradation of the IkappaB-related inhibitor Cactus, liberating Dorsal for nuclear translocation. In addition, other pathways have been suggested to regulate Dorsal. Signaling through the maternal BMP member Decapentaplegic (Dpp) inhibits Dorsal translocation along a pathway parallel to and independent of Toll. In the present study, we show for the first time that the maternal JAK/STAT pathway also regulates embryonic DV patterning. Null alleles of loci coding for elements of the JAK/STAT pathway, hopscotch (hop), marelle (mrl) and zimp (zimp), modify zygotic expression along the DV axis. Genetic analysis suggests that the JAK kinase Hop, most similar to vertebrate JAK2, may modify signals downstream of Dpp. In addition, an activated form of Hop results in increased levels of Cactus and Dorsal proteins, modifying the Dorsal/Cactus ratio and consequently DV patterning. These results indicate that different maternal signals mediated by the Toll, BMP and JAK/STAT pathways may converge to regulate NFkappaB activity in Drosophila.
Collapse
Affiliation(s)
- E S S Lopes
- Departamento de Histologia e Embriologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | |
Collapse
|
26
|
Roth P, Xylourgidis N, Sabri N, Uv A, Fornerod M, Samakovlis C. The Drosophila nucleoporin DNup88 localizes DNup214 and CRM1 on the nuclear envelope and attenuates NES-mediated nuclear export. J Cell Biol 2003; 163:701-6. [PMID: 14638854 PMCID: PMC2173670 DOI: 10.1083/jcb.200304046] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2003] [Accepted: 09/22/2003] [Indexed: 11/22/2022] Open
Abstract
Many cellular responses rely on the control of nucleocytoplasmic transport of transcriptional regulators. The Drosophila nucleoporin Nup88 is selectively required for nuclear accumulation of Rel proteins and full activation of the innate immune response. Here, we investigate the mechanisms underlying its role in nucleocytoplasmic transport. Nuclear import of an nuclear localization signal-enhanced green fluorescent protein (NLS-EGFP) reporter is not affected in DNup88 (members only; mbo) mutants, whereas the level of CRM1-dependent EGFP-nuclear export signal (EGFP-NES) export is increased. We show that the nuclear accumulation of the Drosophila Rel protein Dorsal requires CRM1. DNup88 binds to DNup214 and DCRM1 in vitro, and both proteins become mislocalized from the nuclear rim into the nucleus of mbo mutants. Overexpression of DNup88 is sufficient to relocalize DNup214 and CRM1 on the nuclear envelope and revert the mutant phenotypes. We propose that a major function of DNup88 is to anchor DNup214 and CRM1 on the nuclear envelope and thereby attenuate NES-mediated nuclear export.
Collapse
Affiliation(s)
- Peggy Roth
- Department of Developmental Biology, Wenner-Gren Institute, Stockholm University, S-10691 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
27
|
Maier HJ, Marienfeld R, Wirth T, Baumann B. Critical role of RelB serine 368 for dimerization and p100 stabilization. J Biol Chem 2003; 278:39242-50. [PMID: 12874295 DOI: 10.1074/jbc.m301521200] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In mature B cells RelB-containing complexes are constitutively present in the nucleus, and they are less susceptible to inhibitory kappaB proteins. In most other cell types inhibitory kappaB proteins prevent nuclear translocation and activation of NFkappaB. We reasoned that this characteristic might be because of post-translational modifications of RelB. In Drosophila, signal-dependent phosphorylation of the Rel homologue Dorsal at serine 317 has been shown to be critical for nuclear import. The evolutionary conservation of this serine prompted us to analyze the function of the corresponding site in RelB. As a model system we used the murine S107 plasmacytoma cell line, which lacks endogenous RelB expression. Analysis of S107 cells expressing wild type RelB and serine 368 mutants reveals that serine 368 is not required for nuclear import but that it is critical for RelB dimerization with other members of the NFkappaB family. Similar effects were obtained when the conserved serine in RelA was mutated. We further demonstrate that expression of functional RelB, but not of serine 368 mutants, severely reduces p52 generation and strongly increases expression of the p52 precursor, p100. Wild type RelB, but not mutant RelB, prolonged p100 half-life. We therefore suggest an inhibitory effect of RelB on p100 processing, which is possibly regulated in a signal-dependent manner.
Collapse
Affiliation(s)
- Harald Jakob Maier
- Department of Physiological Chemistry, Ulm University, Ulm 89081, Germany
| | | | | | | |
Collapse
|
28
|
Bolatto C, Chifflet S, Megighian A, Cantera R. Synaptic activity modifies the levels of Dorsal and Cactus at the neuromuscular junction of Drosophila. JOURNAL OF NEUROBIOLOGY 2003; 54:525-36. [PMID: 12532402 DOI: 10.1002/neu.10179] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The Drosophila Rel transcription factor Dorsal and its inhibitor Cactus participate in a signal transduction pathway involved in several biologic processes, including embryonic pattern formation, immunity, and muscle development. In contrast with embryonic muscle, where Dorsal is reportedly absent, this protein and Cactus accumulates in the neuromuscular junctions in the muscle of both larvae and adults. The phenotype of homozygous dorsal mutant larvae suggested that Dorsal and Cactus maybe necessary for normal function and maintenance of the neuromuscular system. Here we investigate if these proteins can respond to synaptic activity. Using larval body wall preparations and antibodies specific for Dorsal or Cactus we show that the amount of these proteins at the neuromuscular junction is substantially decreased after electrical stimulation of the nerves or incubation in glutamate, the principal transmitter in this type of synapse. The specificity of the response was tested with a glutamate receptor antagonist (argiotoxin 636). Because the effect can be reproduced using a calcium ionophore (ionomycin treatment) as well as blocked by the inhibition of the muscle ryanodine receptor (tetracaine treatment), the involvement of calcium in this process seems likely. We also observed that the inhibition of the calcium dependent protein phosphatase calcineurin prevents the effect of glutamate on the fluorescence for Dorsal and Cactus, suggesting its participation in a signal transduction cascade that may activate Dorsal in the muscle independently of Toll. Our results are consistent with a novel function of the Rel factor Dorsal in a molecular pathway turned on by neural activity and/or contractile activity.
Collapse
Affiliation(s)
- Carmen Bolatto
- Departamento de Histología y Embriología, Facultad de Medicina, Universidad de la República, Uruguay
| | | | | | | |
Collapse
|
29
|
Janssens S, Beyaert R. Functional diversity and regulation of different interleukin-1 receptor-associated kinase (IRAK) family members. Mol Cell 2003; 11:293-302. [PMID: 12620219 DOI: 10.1016/s1097-2765(03)00053-4] [Citation(s) in RCA: 444] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Interleukin-1 receptor-associated kinase (IRAK) was first described as a signal transducer for the proinflammatory cytokine interleukin-1 (IL-1) and was later implicated in signal transduction of other members of the Toll-like receptor (TLR)/IL-1 receptor (IL-1R) family. In the meantime, four different IRAK-like molecules have been identified: two active kinases, IRAK-1 and IRAK-4, and two inactive kinases, IRAK-2 and IRAK-M. All IRAKs mediate activation of nuclear factor-kappaB (NF-kappaB) and mitogen-activated protein kinase (MAPK) pathways. Although earlier observations suggested that IRAKs have redundant functions, this hypothesis is now challenged by knockout studies. Furthermore, recent data imply a role for IRAK-1 in tumor necrosis factor receptor (TNFR) superfamily-induced signaling pathways as well. The scope of this review is to highlight the specific role of different IRAKs and to discuss several mechanisms that contribute to their activation and regulation.
Collapse
Affiliation(s)
- Sophie Janssens
- Department of Molecular Biomedical Research, Unit of Molecular Signal Transduction in Inflammation, Ghent University, VIB, K.L. Ledeganckstraat 35, B-9000, Ghent, Belgium
| | | |
Collapse
|
30
|
Kambris Z, Bilak H, D'Alessandro R, Belvin M, Imler JL, Capovilla M. DmMyD88 controls dorsoventral patterning of the Drosophila embryo. EMBO Rep 2003; 4:64-9. [PMID: 12524523 PMCID: PMC1315814 DOI: 10.1038/sj.embor.embor714] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2002] [Revised: 10/13/2002] [Accepted: 11/06/2002] [Indexed: 11/08/2022] Open
Abstract
MyD88 is an adapter protein in the signal transduction pathway mediated by interleukin-1 (IL-1) and Toll-like receptors. A Drosophila homologue of MyD88 (DmMyD88) was recently shown to be required for the Toll-mediated immune response. In Drosophila, the Toll pathway was originally characterized for its role in the dorsoventral patterning of the embryo. We found that, like Toll, DmMyD88 messenger RNA is maternally supplied to the embryo. Here we report the identification of a new mutant allele of DmMyD88, which generates a protein lacking the carboxy-terminal extension, normally located downstream of the Toll/IL-1 receptor domain. Homozygous mutant female flies lay dorsalized embryos that are rescued by expression of a transgenic DmMyD88 complementary DNA. The DmMyD88 mutation blocks the ventralizing activity of a gain-of-function Toll mutation. These results show that DmMyD88 encodes an essential component of the Toll pathway in dorsoventral pattern formation.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing
- Alleles
- Amino Acid Sequence
- Animals
- Animals, Genetically Modified
- Antigens, Differentiation/chemistry
- Antigens, Differentiation/genetics
- Antigens, Differentiation/physiology
- Base Sequence
- DNA Transposable Elements
- DNA, Complementary/genetics
- Drosophila Proteins/chemistry
- Drosophila Proteins/genetics
- Drosophila Proteins/physiology
- Drosophila melanogaster/embryology
- Drosophila melanogaster/genetics
- Drosophila melanogaster/immunology
- Egg Proteins/genetics
- Egg Proteins/physiology
- Embryo, Nonmammalian/ultrastructure
- Exons/genetics
- Female
- Gene Expression Regulation, Developmental
- Genotype
- Immunity, Innate
- Male
- Molecular Sequence Data
- Morphogenesis/genetics
- Mutagenesis, Insertional
- Myeloid Differentiation Factor 88
- Oocytes/metabolism
- Protein Biosynthesis
- Protein Structure, Tertiary
- RNA, Messenger/metabolism
- Receptors, Cell Surface/genetics
- Receptors, Immunologic/chemistry
- Receptors, Immunologic/genetics
- Receptors, Immunologic/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Toll-Like Receptors
- Zygote/metabolism
Collapse
Affiliation(s)
- Zakaria Kambris
- Institut de Biologie Moléculaire et Cellulaire, UPR 9022 du CNRS, 15, rue René Descartes, 67084 Strasbourg Cedex, France
- These authors contributed equally to this work
| | - Hana Bilak
- Institut de Biologie Moléculaire et Cellulaire, UPR 9022 du CNRS, 15, rue René Descartes, 67084 Strasbourg Cedex, France
- These authors contributed equally to this work
| | - Rosalba D'Alessandro
- Institut de Biologie Moléculaire et Cellulaire, UPR 9022 du CNRS, 15, rue René Descartes, 67084 Strasbourg Cedex, France
| | - Marcia Belvin
- Exelixis Inc., P.O. Box 511, South San Francisco, California 98083, USA
| | - Jean-Luc Imler
- Institut de Biologie Moléculaire et Cellulaire, UPR 9022 du CNRS, 15, rue René Descartes, 67084 Strasbourg Cedex, France
| | - Maria Capovilla
- Institut de Biologie Moléculaire et Cellulaire, UPR 9022 du CNRS, 15, rue René Descartes, 67084 Strasbourg Cedex, France
- Tel: +33 3 88 41 70 21; Fax: +33 3 88 60 69 22;
| |
Collapse
|
31
|
Shen B, Manley JL. Pelle kinase is activated by autophosphorylation during Toll signaling in Drosophila. Development 2002; 129:1925-33. [PMID: 11934858 DOI: 10.1242/dev.129.8.1925] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The Drosophila Pelle kinase plays a key role in the evolutionarily conserved Toll signaling pathway, but the mechanism responsible for its activation has been unknown. We present in vivo and in vitro evidence establishing an important role for concentration-dependent autophosphorylation in the signaling process. We first show that Pelle phosphorylation can be detected transiently in early embryos, concomitant with activation of signaling. Importantly, Pelle phosphorylation is enhanced in a gain-of-function Toll mutant (Toll10b), but decreased by loss-of-function Toll alleles. Next we found that Pelle is phosphorylated in transfected Schneider L2 cells in a concentration-dependent manner such that significant modification is observed only at high Pelle concentrations, which coincide with levels required for phosphorylation and activation of the downstream target, Dorsal. Pelle phosphorylation is also enhanced in L2 cells co-expressing Toll10b, and is dependent on Pelle kinase activity. In vitro kinase assays revealed that recombinant, autophosphorylated Pelle is far more active than unphosphorylated Pelle. Importantly, unphosphorylated Pelle becomes autophosphorylated, and activated, by incubation at high concentrations. We discuss these results in the context of Toll-like receptor mediated signaling in both flies and mammals.
Collapse
Affiliation(s)
- Baohe Shen
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | |
Collapse
|
32
|
Fernandez NQ, Grosshans J, Goltz JS, Stein D. Separable and redundant regulatory determinants in Cactus mediate its dorsal group dependent degradation. Development 2001; 128:2963-74. [PMID: 11532919 DOI: 10.1242/dev.128.15.2963] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Dorsal-ventral polarity within the Drosophila syncytial blastoderm embryo is determined by the maternally encoded dorsal group signal transduction pathway that regulates nuclear localization of the transcription factor Dorsal. Nuclear uptake of Dorsal, a Rel/NFκB homolog, is controlled by the interaction with its cognate IκB inhibitor protein Cactus, which is degraded on the ventral side of the embryo in response to dorsal group signaling. Previous studies have suggested that an N-terminally located kinase target motif similar to that found in IκB proteins is involved in the spatially controlled degradation of Cactus. We report studies of the in vivo function and distribution of fusion proteins comprising segments of Cactus attached to Escherichia coli β-galactosidase (lacZ). Full-length Cactus-lacZ expressed in vivo normalizes the ventralized phenotype of embryos that lack Cactus and faithfully reconstitutes dorsal group-regulated degradation, while fusion protein constructs that lack the first 125 amino acids of Cactus escape dorsal group-dependent degradation. Furthermore, Cactus-lacZ constructs that lack only the putative IκB-dependent kinase target-like motif can nevertheless undergo spatially regulated dorsal group-dependent degradation and we have identified the regulatory determinant responsible for dorsal group-dependent degradation of Cactus in the absence of this motif. Taken together, our studies indicate the presence of two distinct redundantly acting determinants in the N terminus of Cactus that direct dorsal group-dependent degradation. Strikingly, the regulatory domain of human IκBα can also direct polarized degradation of Cactus-lacZ fusion protein.
Collapse
Affiliation(s)
- N Q Fernandez
- Department of Molecular Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
33
|
Abstract
Protein-protein interactions mediate many important cellular processes and are central to the mechanisms by which most proteins function. Charting the interactions among the proteins involved in a process has been an essential step in characterising the function of proteins and pathways. The yeast two-hybrid system is one approach to detecting protein interactions that can now be scaled-up to assay large sets of proteins systematically, such as those being identified from genome sequencing efforts. The system has already been extensively used to acquire data that have enabled construction of large protein interaction maps (PIMs). When combined with other data, including data being generated by other functional genomics approaches, PIMs help assign function to new proteins and delineate functional networks. Hypotheses generated in such a manner often must be tested by additional experimentation, preferably in vivo. The model organism Drosophila melanogaster has a wealth of genetic and bioinformatic tools available for such analyses. The proteome predicted from the recently sequenced Drosophila genome indicates that humans have more genes in common with Drosophila than with any other invertebrate model organism characterised to date. Thus, the construction and characterisation of Drosophila PIMs will help define the functions of many conserved genes and pathways, and will provide the pharmaceutical research industry with invaluable data to assist with drug target identification and validation.
Collapse
Affiliation(s)
- C A Stanyon
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, 540 East Canfield Avenue, Detroit, Michigan 48201, USA
| | | |
Collapse
|
34
|
Abstract
Experiments reported in the past year have revealed considerable diversity in Toll-mediated pathways for signal transduction in development and innate immunity. Rather than function as a well conserved signaling cassette, Toll receptors and associated factors have apparently evolved as a diverse set of configurations to defend against microbial infection in species ranging from plants to humans.
Collapse
Affiliation(s)
- S A Wasserman
- Center for Molecular Genetics, Division of Biology, University of California, San Diego, 9500 Gilman Drive, MC 0634, La Jolla, California 92093-0634, USA.
| |
Collapse
|
35
|
Uv AE, Roth P, Xylourgidis N, Wickberg A, Cantera R, Samakovlis C. members only encodes a Drosophila nucleoporin required for rel protein import and immune response activation. Genes Dev 2000; 14:1945-57. [PMID: 10921908 PMCID: PMC316830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2000] [Accepted: 06/05/2000] [Indexed: 02/17/2023]
Abstract
Many developmental and physiological responses rely on the selective translocation of transcriptional regulators in and out of the nucleus through the nuclear pores. Here we describe the Drosophila gene members only (mbo) encoding a nucleoporin homologous to the mammalian Nup88. The phenotypes of mbo mutants and mbo expression during development are cell specific, indicating that the nuclear import capacity of cells is differentially regulated. Using inducible assays for nucleocytoplasmic trafficking we show that mRNA export and classic NLS-mediated protein import are unaffected in mbo mutants. Instead, mbo is selectively required for the nuclear import of the yeast transcription factor GAL4 in a subset of the larval tissues. We have identified the first endogenous targets of the mbo nuclear import pathway in the Rel proteins Dorsal and Dif. In mbo mutants the upstream signaling events leading to the degradation of the IkappaB homolog Cactus are functional, but Dorsal and Dif remain cytoplasmic and the larval immune response is not activated in response to infection. Our results demonstrate that distinct nuclear import events require different nucleoporins in vivo and suggest a regulatory role for mbo in signal transduction.
Collapse
Affiliation(s)
- A E Uv
- Umeâ Center for Molecular Pathogenesis (UCMP), Umeâ University, S-90187 Umeâ, Sweden
| | | | | | | | | | | |
Collapse
|
36
|
Olesen CE, Mosier J, Voyta JC, Bronstein I. Chemiluminescent immunodetection protocols with 1,2-dioxetane substrates. 4. Methods Enzymol 2000; 305:417-27. [PMID: 10812617 DOI: 10.1016/s0076-6879(00)05504-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Chemiluminescent 1,2-dioxetane enzyme substrates provide a highly sensitive and versatile detection method for immunoblots and other membrane-based detections. 1,2-Dioxetane substrates, coupled with either alkaline phosphatase or beta-galactosidase enzyme labels, generate glow light emission kinetics, with a signal duration that is significantly longer than most enhanced luminol/horseradish peroxidase chemiluminescent detection systems. The long-lived, high-intensity light signal is ideal for imaging using a variety of formats, including X-ray film, photographic film, chemiluminescence phosphor imaging screens, and the rapidly expanding selection of camera imaging systems.
Collapse
Affiliation(s)
- C E Olesen
- Tropix, Inc., Bedford, Massachusetts 01730, USA
| | | | | | | |
Collapse
|
37
|
Gordon EH, Page MD, Willis AC, Ferguson SJ. Escherichia coli DipZ: anatomy of a transmembrane protein disulphide reductase in which three pairs of cysteine residues, one in each of three domains, contribute differentially to function. Mol Microbiol 2000; 35:1360-74. [PMID: 10760137 DOI: 10.1046/j.1365-2958.2000.01796.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
DipZ is a bacterial cytoplasmic membrane protein that transfers reducing power from the cytoplasm to the periplasm so as to facilitate the formation of correct disulphide bonds and c-type cytochromes in the latter compartment. Topological analysis using gene fusions between the Escherichia coli dipZ and either E. coli phoA or lacZ shows that DipZ has a highly hydrophobic central domain comprising eight transmembrane alpha-helices plus periplasmic globular N-terminal and C-terminal domains. The previously assigned translational start codon for the E. coli DipZ was shown to be incorrect and the protein to be larger than previously thought. The experimentally determined translational start position indicates that an additional alpha-helix at the N-terminus acts as a cleavable signal peptide so that the N-terminus of the mature protein is located in the periplasm. The newly assigned 5' end of the dipZ gene was shown to be preceded by a functional ribosome-binding site. The hydrophobic central domain and both of the periplasmic globular domains each have a pair of highly conserved cysteine residues, and it was shown by site directed mutagenesis that all six conserved cysteine residues contribute to DipZ function.
Collapse
Affiliation(s)
- E H Gordon
- Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | | | | | | |
Collapse
|
38
|
Drier EA, Govind S, Steward R. Cactus-independent regulation of Dorsal nuclear import by the ventral signal. Curr Biol 2000; 10:23-6. [PMID: 10660298 DOI: 10.1016/s0960-9822(99)00267-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Rel-family transcription factors function in a variety of biological processes, including development and immunity. During early Drosophila development, the Toll-Cactus-Dorsal pathway regulates the establishment of the embryonic dorsoventral axis. The last step in this pathway is the graded nuclear import of the Rel protein Dorsal. Dorsal is retained in the cytoplasm by the IkappaB-family protein Cactus. Phosphorylation of both Dorsal and Cactus is regulated by a Toll-receptor-dependent ventral signal relayed by the Tube and Pelle proteins. Phosphorylation of Cactus leads to its degradation and to the release of Dorsal to form a ventral-to-dorsal nuclear Dorsal gradient. To understand how the ventral signal regulates the nuclear import and activity of Dorsal, we deleted its conserved nuclear localization signal (NLS). The truncated protein remained in the cytoplasm and could antagonize the function of wild-type Dorsal, suggesting that Dorsal forms a dimer in the cytoplasm. Further, the nuclear import of a mutant Dorsal protein that failed to interact with Cactus was still regulated by the ventral signal. Our results are consistent with a model in which ventral signal-dependent modification of both Cactus and Dorsal is required for the graded nuclear import of Dorsal.
Collapse
Affiliation(s)
- E A Drier
- Waksman Institute, Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, New Jersey 08854-8020, USA
| | | | | |
Collapse
|
39
|
Xiao T, Towb P, Wasserman SA, Sprang SR. Three-dimensional structure of a complex between the death domains of Pelle and Tube. Cell 1999; 99:545-55. [PMID: 10589682 PMCID: PMC4372121 DOI: 10.1016/s0092-8674(00)81542-1] [Citation(s) in RCA: 144] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The interaction of the serine/threonine kinase Pelle and adaptor protein Tube through their N-terminal death domains leads to the nuclear translocation of the transcription factor Dorsal and activation of zygotic patterning genes during Drosophila embryogenesis. Crystal structure of the Pelle and Tube death domain heterodimer reveals that the two death domains adopt a six-helix bundle fold and are arranged in an open-ended linear array with plastic interfaces mediating their interactions. The Tube death domain has an insertion between helices 2 and 3, and a C-terminal tail making significant and indispensable contacts in the heterodimer. In vivo assays of Pelle and Tube mutants confirmed that the integrity of the major heterodimer interface is critical to the activity of these molecules.
Collapse
Affiliation(s)
- Tsan Xiao
- The Howard Hughes Medical Institute and Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75235-9050
| | - Par Towb
- Department of Biology, University of California at San Diego, La Jolla, California 92093-0634
| | - Steven A. Wasserman
- Department of Biology, University of California at San Diego, La Jolla, California 92093-0634
| | - Stephen R. Sprang
- The Howard Hughes Medical Institute and Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas 75235-9050
- To whom correspondence should be addressed ()
| |
Collapse
|
40
|
Abstract
The Drosophila Rel/NF-kappaB transcription factors - Dorsal, Dif, and Relish - control several biological processes, including embryonic pattern formation, muscle development, immunity, and hematopoiesis. Molecular-genetic analysis of 12 mutations that cause embryonic dorsal/ventral patterning defects has defined the steps that control the formation of this axis. Regulated activation of the Toll receptor leads to the establishment of a gradient of nuclear Dorsal protein, which in turn governs the subdivision of the axis and specification of ventral, lateral and dorsal fates. Phenotypic analysis of dorsal-ventral embryonic mutants and the characterization of the two other fly Rel proteins, Dif and Relish, have shown that the intracellular portion of the Toll to Cactus pathway also controls the innate immune response in Drosophila. Innate immunity and hematopoiesis are regulated by analogous Rel/NF-kappaB-family pathways in mammals. The elucidation of the complex regulation and diverse functions of Drosophila Rel proteins underscores the relevance of basic studies in Drosophila.
Collapse
Affiliation(s)
- S Govind
- Department of Biology, City College and The Graduate Center of The City University of New York, 138th Street and Convent Avenue, New York, NY 10031, USA
| |
Collapse
|
41
|
Engström Y. Induction and regulation of antimicrobial peptides in Drosophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 1999; 23:345-358. [PMID: 10426427 DOI: 10.1016/s0145-305x(99)00016-6] [Citation(s) in RCA: 114] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Activation of the innate immune response involves recognition of the infectious agent and the subsequent activation of cellular and humoral reactions. In insects, a number of immunity genes are activated at the level of transcription leading to the synthesis of antimicrobial peptides. Genetic analyses in Drosophila have identified several signal transduction pathways that promote activation of these immunity genes. Recent data suggest that the insect immune system is able to discriminate between a bacterial and a fungal infection, and responds by higher levels of activation of the appropriate peptides to repel the infection. These and other recent data on transcription factors and regulation of antimicrobial genes are integrated into a model to suggest how differential activation of antifungal and antibacterial peptides can occur in response to fungal and bacterial infection.
Collapse
Affiliation(s)
- Y Engström
- Department of Molecular Biology, Stockholm University, Sweden.
| |
Collapse
|
42
|
|
43
|
Cheng MH, Maines JZ, Wasserman SA. Biphasic subcellular localization of the DAZL-related protein boule in Drosophila spermatogenesis. Dev Biol 1998; 204:567-76. [PMID: 9882490 DOI: 10.1006/dbio.1998.9098] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The Drosophila boule gene is expressed exclusively in the male germline and encodes an RNA binding protein closely related to the mammalian fertility factors encoded by the DAZ (Deleted in Azoospermia) and DAZL (DAZ-like) genes. Mutation of boule blocks both meiotic divisions. Differentiation nonetheless continues, resulting in tetraploid spermatids that fail to mature into sperm. We have found that Boule localizes premeiotically to a perinucleolar region and then translocates to the cytoplasm at the onset of meiosis. We show that deletion of the Y chromosome ks-1 fertility locus eliminates Boule nuclear localization, although it does not perturb entry into meiosis. Based on these observations we propose that Boule acts in the cytoplasm to regulate the stability or translation of messenger RNA encoding an essential meiotic factor.
Collapse
Affiliation(s)
- M H Cheng
- Department of Molecular Biology and Oncology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas, 75235-9148, USA
| | | | | |
Collapse
|
44
|
Shen B, Manley JL. Phosphorylation modulates direct interactions between the Toll receptor, Pelle kinase and Tube. Development 1998; 125:4719-28. [PMID: 9806920 DOI: 10.1242/dev.125.23.4719] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Determination of dorsal/ventral polarity in Drosophila requires 12 genetically defined, maternally encoded proteins. These include Toll, a transmembrane receptor, Pelle, a ser/thr protein kinase and Tube, all of which function intracytoplasmically to initiate the cascade that ultimately activates Dorsal, an NF-kappaB family transcription factor. Here we describe biochemical interactions between recombinant Toll, Pelle and Tube that provide insights into early events in activation of the signaling cascade. We first show that Pelle binds directly to a region within the Toll intracytoplasmic domain, providing the first evidence that these two evolutionarily conserved molecules physically interact. We then demonstrate that Pelle can be autophosphorylated, and that this prevents binding to Toll as well as Tube. Autophosphorylation occurs in the N-terminal, death-domain-containing region of Pelle, which is dispensable for binding to Toll but required for enzymatic activity. We also show that Pelle phosphorylates Toll, within the region required for Pelle interaction, but this phosphorylation can be blocked by a previously characterized inhibitory domain at the Toll C terminus. These and other results allow us to propose a model by which multiple phosphorylation-regulated interactions between these three proteins lead to activation of the Dorsal signaling pathway.
Collapse
Affiliation(s)
- B Shen
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | | |
Collapse
|
45
|
Briggs LJ, Stein D, Goltz J, Corrigan VC, Efthymiadis A, Hübner S, Jans DA. The cAMP-dependent protein kinase site (Ser312) enhances dorsal nuclear import through facilitating nuclear localization sequence/importin interaction. J Biol Chem 1998; 273:22745-52. [PMID: 9712906 DOI: 10.1074/jbc.273.35.22745] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Control over the nuclear import of transcription factors (TFs) represents a level of gene regulation integral to cellular processes such as differentiation and transformation. The Drosophila TF Dorsal shares with other rel TF family members the fact that it contains a phosphorylation site for the cAMP-dependent protein kinase (PKA) 22 amino acids N-terminal to the nuclear localization signal (NLS) at amino acids 335-340. This study examines for the first time the nuclear import kinetics of Dorsal fusion proteins in rat hepatoma cells in vivo and in vitro. Nuclear uptake was found to be not only NLS-dependent, but also strongly dependent on the PKA site, whereby substitution of Ser312 by either Ala or Glu using site-directed mutagenesis severely reduced nuclear accumulation. Exogenous cAMP or PKA catalytic subunit significantly enhanced the nuclear import of wild-type proteins both in vivo and in vitro. Using a direct binding assay, the molecular basis of PKA site enhancement of Dorsal fusion protein nuclear import was determined to be PKA site-mediated modulation of NLS recognition by the importin 58/97 complex. The physiological relevance of these results is supported by the observation that Drosophila embryos expressing PKA site Dorsal mutant variants were impaired in development. We conclude that the Dorsal NLS and PKA site constitute a phosphorylation-regulated NLS essential to Dorsal function and able to function in heterologous mammalian cell systems, where phosphorylation modulates the affinity of NLS recognition by importin.
Collapse
Affiliation(s)
- L J Briggs
- Nuclear Signaling Laboratory, Division of Biochemistry and Molecular Biology, John Curtin School of Medical Research, Canberra City, A.C. T. 2601, Australia
| | | | | | | | | | | | | |
Collapse
|
46
|
Stein D, Goltz JS, Jurcsak J, Stevens L. The Dorsal-related immunity factor (Dif) can define the dorsal-ventral axis of polarity in the Drosophila embryo. Development 1998; 125:2159-69. [PMID: 9570779 DOI: 10.1242/dev.125.11.2159] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In Drosophila embryos, dorsal-ventral polarity is defined by a signal transduction pathway that regulates nuclear import of the Dorsal protein. Dorsal protein's ability to act as a transcriptional activator of some zygotic genes and a repressor of others defines structure along the dorsal-ventral axis. Dorsal is a member of a group of proteins, the Rel-homologous proteins, whose activity is regulated at the level of nuclear localization. Dif, a more recently identified Drosophila Rel-homologue, has been proposed to act as a mediator of the immune response in Drosophila. In an effort to understand the function and regulation of Rel-homologous proteins in Drosophila, we have expressed Dif protein in Drosophila embryos derived from dorsal mutant mothers. We found that the Dif protein was capable of restoring embryonic dorsal-ventral pattern elements and was able to define polarity correctly with respect to the orientation of the egg shell. This, together with the observation that the ability of Dif to restore a dorsal-ventral axis depended on the signal transduction pathway that normally regulates Dorsal, suggests that Dif protein formed a nuclear concentration gradient similar to that seen for Dorsal. By studying the expression of Dorsal target genes we found that Dif could activate the zygotic genes that Dorsal activates and repress the genes repressed by Dorsal. Differences in the expression of these target genes, as well as the results from interaction studies carried out in yeast, suggest that Dif is not capable of synergizing with the basic helix-loop-helix transcription factors with which Dorsal normally interacts, and thereby lacks an important component of Dorsal-mediated pattern formation.
Collapse
Affiliation(s)
- D Stein
- Department of Molecular Genetics and Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA.
| | | | | | | |
Collapse
|
47
|
Ghosh S, May MJ, Kopp EB. NF-kappa B and Rel proteins: evolutionarily conserved mediators of immune responses. Annu Rev Immunol 1998; 16:225-60. [PMID: 9597130 DOI: 10.1146/annurev.immunol.16.1.225] [Citation(s) in RCA: 4129] [Impact Index Per Article: 152.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The transcription factor NF-kappa B, more than a decade after its discovery, remains an exciting and active area of study. The involvement of NF-kappa B in the expression of numerous cytokines and adhesion molecules has supported its role as an evolutionarily conserved coordinating element in the organism's response to situations of infection, stress, and injury. Recently, significant advances have been made in elucidating the details of the pathways through which signals are transmitted to the NF-kappa B:I kappa B complex in the cytosol. The field now awaits the discovery and characterization of the kinase responsible for the inducible phosphorylation of I kappa B proteins. Another exciting development has been the demonstration that in certain situations NF-kappa B acts as an anti-apoptotic protein; therefore, elucidation of the mechanism by which NF-kappa B protects against cell death is an important goal. Finally, the generation of knockouts of members of the NF-kappa B/I kappa B family has allowed the study of the roles of these proteins in normal development and physiology. In this review, we discuss some of these recent findings and their implications for the study of NF-kappa B.
Collapse
Affiliation(s)
- S Ghosh
- Section of Immunobiology, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06520, USA.
| | | | | |
Collapse
|
48
|
Viehweg J, Naumann WW, Olsson R. Secretory Radial Glia in the Ectoneural System of the Sea StarAsterias rubens(Echinodermata). ACTA ZOOL-STOCKHOLM 1998. [DOI: 10.1111/j.1463-6395.1998.tb01151.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
49
|
Edwards DN, Towb P, Wasserman SA. An activity-dependent network of interactions links the Rel protein Dorsal with its cytoplasmic regulators. Development 1997; 124:3855-64. [PMID: 9367441 DOI: 10.1242/dev.124.19.3855] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A signaling pathway active on the ventral side of the Drosophila embryo defines dorsoventral polarity. A cell surface signal relayed by Toll, Tube and Pelle releases the Rel-related protein Dorsal from its cytoplasmic inhibitor Cactus; free Dorsal translocates into nuclei and directs expression of ventral fates. Using the yeast two-hybrid system and immunoprecipitation experiments, we define scaffolding and anchoring interactions among the pathway components. We show that Dorsal binds specifically to Tube, Pelle and Cactus, and that the protein kinase activity of Pelle differentially regulates its interactions with Dorsal and Tube. We also identify Drosophila Filamin as a potential adaptor linking the interaction network, via Tube, to the transmembrane receptor Toll.
Collapse
Affiliation(s)
- D N Edwards
- Department of Molecular Biology and Oncology, UT Southwestern Medical Center, Dallas, Texas 75235-9148, USA
| | | | | |
Collapse
|
50
|
Zhong H, SuYang H, Erdjument-Bromage H, Tempst P, Ghosh S. The transcriptional activity of NF-kappaB is regulated by the IkappaB-associated PKAc subunit through a cyclic AMP-independent mechanism. Cell 1997; 89:413-24. [PMID: 9150141 DOI: 10.1016/s0092-8674(00)80222-6] [Citation(s) in RCA: 677] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Stimulation of cells with inducers of NF-kappaB such as LPS and IL-1 leads to the degradation of IkappaB-alpha and IkappaB-beta proteins and translocation of NF-kappaB to the nucleus. We now demonstrate that, besides the physical partitioning of inactive NF-kappaB to the cytosol, the transcriptional activity of NF-kappaB is regulated through phosphorylation of NF-kappaB p65 by protein kinase A (PKA). The catalytic subunit of PKA (PKAc) is maintained in an inactive state through association with IkappaB-alpha or IkappaB-beta in an NF-kappaB-IkappaB-PKAc complex. Signals that cause the degradation of IkappaB result in activation of PKAc in a cAMP-independent manner and the subsequent phosphorylation of p65. Therefore, this pathway represents a novel mechanism for the cAMP-independent activation of PKA and the regulation of NF-kappaB activity.
Collapse
Affiliation(s)
- H Zhong
- Department of Molecular Biophysics and Biochemistry, Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | | | | | | | |
Collapse
|