51
|
Chen L, Paquette N, Mamoor S, Rus F, Nandy A, Leszyk J, Shaffer SA, Silverman N. Innate immune signaling in Drosophila is regulated by transforming growth factor β (TGFβ)-activated kinase (Tak1)-triggered ubiquitin editing. J Biol Chem 2017; 292:8738-8749. [PMID: 28377500 DOI: 10.1074/jbc.m117.788158] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Indexed: 11/06/2022] Open
Abstract
Coordinated regulation of innate immune responses is necessary in all metazoans. In Drosophila the Imd pathway detects Gram-negative bacterial infections through recognition of diaminopimelic acid (DAP)-type peptidoglycan and activation of the NF-κB precursor Relish, which drives robust antimicrobial peptide gene expression. Imd is a receptor-proximal adaptor protein homologous to mammalian RIP1 that is regulated by proteolytic cleavage and Lys-63-polyubiquitination. However, the precise events and molecular mechanisms that control the post-translational modification of Imd remain unclear. Here, we demonstrate that Imd is rapidly Lys-63-polyubiquitinated at lysine residues 137 and 153 by the sequential action of two E2 enzymes, Ubc5 and Ubc13-Uev1a, in conjunction with the E3 ligase Diap2. Lys-63-ubiquitination activates the TGFβ-activated kinase (Tak1), which feeds back to phosphorylate Imd, triggering the removal of Lys-63 chains and the addition of Lys-48 polyubiquitin. This ubiquitin-editing process results in the proteasomal degradation of Imd, which we propose functions to restore homeostasis to the Drosophila immune response.
Collapse
Affiliation(s)
- Li Chen
- From the Division of Infectious Disease, Department of Medicine and
| | | | - Shahan Mamoor
- From the Division of Infectious Disease, Department of Medicine and
| | - Florentina Rus
- From the Division of Infectious Disease, Department of Medicine and
| | - Anubhab Nandy
- From the Division of Infectious Disease, Department of Medicine and
| | - John Leszyk
- the Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Scott A Shaffer
- the Proteomics and Mass Spectrometry Facility, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Neal Silverman
- From the Division of Infectious Disease, Department of Medicine and
| |
Collapse
|
52
|
Inhibition of a NF-κB/Diap1 Pathway by PGRP-LF Is Required for Proper Apoptosis during Drosophila Development. PLoS Genet 2017; 13:e1006569. [PMID: 28085885 PMCID: PMC5279808 DOI: 10.1371/journal.pgen.1006569] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 01/30/2017] [Accepted: 01/04/2017] [Indexed: 12/15/2022] Open
Abstract
NF-κB pathways are key signaling cascades of the Drosophila innate immune response. One of them, the Immune Deficiency (IMD) pathway, is under a very tight negative control. Although molecular brakes exist at each step of this signaling module from ligand availability to transcriptional regulation, it remains unknown whether repressors act in the same cells or tissues and if not, what is rationale behind this spatial specificity. We show here that the negative regulator of IMD pathway PGRP-LF is epressed in ectodermal derivatives. We provide evidence that, in the absence of any immune elicitor, PGRP-LF loss-of-function mutants, display a constitutive NF-κB/IMD activation specifically in ectodermal tissues leading to genitalia and tergite malformations. In agreement with previous data showing that proper development of these structures requires induction of apoptosis, we show that ectopic activation of NF-κB/IMD signaling leads to apoptosis inhibition in both genitalia and tergite primordia. We demonstrate that NF-κB/IMD signaling antagonizes apoptosis by up-regulating expression of the anti-apoptotic protein Diap1. Altogether these results show that, in the complete absence of infection, the negative regulation of NF-κB/IMD pathway by PGRP-LF is crucial to ensure proper induction of apoptosis and consequently normal fly development. These results highlight that IMD pathway regulation is controlled independently in different tissues, probably reflecting the different roles of this signaling cascade in both developmental and immune processes. In multicellular organism such as mammals or insects, activation of innate immune responses occurs following detection of microbes by dedicated receptors called pattern recognition receptors. Such immune activation is taking place in immune competent tissue such as the skin, the digestive and respiratory epithelia and is under a tight negative control. Negative control is essential to finely adjust the duration and the intensity of the immune response to the level of infection. We found that the Drosophila innate immunity negative regulator PGRP-LF, is specifically expressed in non-immune tissues and plays an essential role during development, in absence of any infection. Lack of PGRP-LF function in these tissues inhibits apoptosis leading to incomplete genitalia rotation and tergite malformations. We show that such apoptosis inhibition results from the over expression of the negative regulator of apoptosis Diap1 specifically in PGRP-LF expressing cells. Our data highlight that proper negative regulation of immune signaling pathway in non-immune tissues is contributing to normal development and illustrate the growing evidence of the dual role of immune signaling pathway contribution to both immunity and in development processes.
Collapse
|
53
|
Tanaka H, Sagisaka A. Identification and functional analysis of pointed homologs in Bombyx mori. Gene 2016; 604:22-32. [PMID: 27988233 DOI: 10.1016/j.gene.2016.12.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/29/2016] [Accepted: 12/12/2016] [Indexed: 12/18/2022]
Abstract
Using gene-knockdown techniques, we searched for endogenous Ets family proteins involved in the regulation of Escherichia coli-dependent lebocin promoter activation in the E. coli-responsive silkworm cell line NIAS-Bm-aff3. Results showed that the gene knockdown of BmPointeds (BmPNTs), Drosophila Pointed orthologs, enhanced E. coli-dependent lebocin promoter activation, suggesting that endogenous BmPNTs repress the activation of this promoter. Furthermore, we found that i) the BmPNT gene produced at least two alternative splicing isoforms, BmPNT1 and BmPNT2, both of which function as repressors; ii) BmPNTs were not associated with an already-reported repressor element, most proximal GGAA/T motif (EtsRE3), in lebocin promoter, which plays a role in the repression of E. coli- and BmRelish1-dependent lebocin promoter activation; iii) although BmPNTs did not directly affect BmRelish1-dependent lebocin promoter activation, they were able to directly repress its activation on the promoter lacking EtsRE3, probably because of competitive inhibition of binding of BmRelish1 to κB sites by BmPNTs; and iv) BmPNTs were mainly expressed in larval hemocytes, and the gene expression levels of BmPNT2, but not of BmPNT1, were decreased in response to E. coli and Bacillus subtilis. These findings suggest that endogenous BmPNTs are directly and indirectly involved in the repression of E. coli-mediated lebocin promoter activation in NIAS-Bm-aff3 cells.
Collapse
Affiliation(s)
- Hiromitsu Tanaka
- Insect-Microbe Research Unit, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba 305-8634, Japan.
| | - Aki Sagisaka
- Insect-Microbe Research Unit, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba 305-8634, Japan
| |
Collapse
|
54
|
Xiong XP, Kurthkoti K, Chang KY, Li JL, Ren X, Ni JQ, Rana TM, Zhou R. miR-34 Modulates Innate Immunity and Ecdysone Signaling in Drosophila. PLoS Pathog 2016; 12:e1006034. [PMID: 27893816 PMCID: PMC5125713 DOI: 10.1371/journal.ppat.1006034] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 10/31/2016] [Indexed: 12/31/2022] Open
Abstract
microRNAs are endogenous small regulatory RNAs that modulate myriad biological processes by repressing target gene expression in a sequence-specific manner. Here we show that the conserved miRNA miR-34 regulates innate immunity and ecdysone signaling in Drosophila. miR-34 over-expression activates antibacterial innate immunity signaling both in cultured cells and in vivo, and flies over-expressing miR-34 display improved survival and pathogen clearance upon Gram-negative bacterial infection; whereas miR-34 knockout animals are defective in antibacterial defense. In particular, miR-34 achieves its immune-stimulatory function, at least in part, by repressing the two novel target genes Dlg1 and Eip75B. In addition, our study reveals a mutual repression between miR-34 expression and ecdysone signaling, and identifies miR-34 as a node in the intricate interplay between ecdysone signaling and innate immunity. Lastly, we identify cis-regulatory genomic elements and trans-acting transcription factors required for optimal ecdysone-mediated repression of miR-34. Taken together, our study enriches the repertoire of immune-modulating miRNAs in animals, and provides new insights into the interplay between steroid hormone signaling and innate immunity.
Collapse
Affiliation(s)
- Xiao-Peng Xiong
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
| | - Krishna Kurthkoti
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
| | - Kung-Yen Chang
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Department of Pediatrics, University of California San Diego School of Medicine, California, United States of America
| | - Jian-Liang Li
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida, United States of America
| | - Xingjie Ren
- Gene Regulatory Laboratory, School of Medicine, Tsinghua University, Beijing, China
| | - Jian-Quan Ni
- Gene Regulatory Laboratory, School of Medicine, Tsinghua University, Beijing, China
| | - Tariq M. Rana
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Department of Pediatrics, University of California San Diego School of Medicine, California, United States of America
| | - Rui Zhou
- Tumor Initiation and Maintenance Program; Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, California, United States of America
- * E-mail:
| |
Collapse
|
55
|
Lamiable O, Meignin C, Imler JL. WntD and Diedel: Two immunomodulatory cytokines in Drosophila immunity. Fly (Austin) 2016; 10:187-94. [PMID: 27314646 DOI: 10.1080/19336934.2016.1202387] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Remarkable progress has been made on the understanding of the basic mechanisms of innate immunity in flies, from sensing infection to production of effector molecules. However, how the immune response is orchestrated at the level of the organism remains poorly understood. While cytokines activating immune responses, such as Spaetzle or Unpaired-3, have been identified and characterized in Drosophila, much less is known regarding immunosuppressor cytokines. In a recent publication, we reported the identification of a novel cytokine, Diedel, which acts as systemic negative regulator of the IMD pathway. Here, we discuss the similarities between Diedel and WntD, another immunomodulatory cytokine and present evidence that the 2 molecules act independently from one another.
Collapse
Affiliation(s)
- Olivier Lamiable
- a CNRS UPR9022 , Institut de Biologie Moléculaire et Cellulaire , Strasbourg , France
| | - Carine Meignin
- a CNRS UPR9022 , Institut de Biologie Moléculaire et Cellulaire , Strasbourg , France.,b Faculté des Sciences de la Vie , Université de Strasbourg , Strasbourg , France
| | - Jean-Luc Imler
- a CNRS UPR9022 , Institut de Biologie Moléculaire et Cellulaire , Strasbourg , France.,b Faculté des Sciences de la Vie , Université de Strasbourg , Strasbourg , France
| |
Collapse
|
56
|
Li X, Meng K, Qiao J, Liu H, Zhong C, Liu Q. Identification of Aadnr1, a novel gene related to innate immunity and apoptosis in Aedes albopictus. Gene 2016; 587:18-26. [PMID: 27045774 DOI: 10.1016/j.gene.2016.03.046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 03/24/2016] [Accepted: 03/25/2016] [Indexed: 12/01/2022]
Abstract
Innate immunity and apoptosis play critical roles in defending pathogens in insects. In Drosophila, Dnr1 was reported as a negative regulator of apoptosis and immune deficiency (Imd) pathway which belongs to innate immunity. Aedes albopictus is an important kind of arbovirus vector and becoming a significant threat to public health due to its rapid global expansion. Here we identified an ortholog of dnr1 from A. albopictus, named as Aadnr1. Aadnr1 encoded a putative protein containing an N-terminal FERM domain and a C-terminal RING domain. AaDnr1 shared high identity with dipteran insects Dnr1 orthologs. Phylogenetic analyses showed that the closest relative of AaDnr1 was Aedes aegypti Dnr1. Real-time PCR proved that Aadnr1 mRNA was expressed ubiquitously during developmental and adult stages. Transcriptional levels of Aadnr1 were decreased drastically in C6/36 cells underwent apoptosis induced by Actinomycin D (Act D) treatment. Partial silence of Aadnr1 enhanced Act D-induced caspase activity. When challenged by heat-inactivated E. coli, transcriptional level of Aadnr1 was also decreased dramatically in C6/36 cells. While when C6/36 cells were infected with Sindbis virus TE/GFP, transcriptional level of Aadnr1 was reduced and recovered repeatedly, with an overall decreasing trend. It was also shown in this study that similar to Drosophila Dnr1, RING domain destabilized AaDnr1 protein. Taken together, the study identified an innate immunity and apoptosis related gene Aadnr1 in A. albopictus.
Collapse
Affiliation(s)
- Xiaomei Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Kun Meng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Jialu Qiao
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Hao Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Chunyan Zhong
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China
| | - Qingzhen Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, People's Republic of China.
| |
Collapse
|
57
|
Mosquito C-type lectins maintain gut microbiome homeostasis. Nat Microbiol 2016; 1:16023. [PMID: 27572642 DOI: 10.1038/nmicrobiol.2016.23] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/01/2016] [Indexed: 12/18/2022]
Abstract
The long-term evolutionary interaction between the host immune system and symbiotic bacteria determines their cooperative rather than antagonistic relationship. It is known that commensal bacteria have evolved a number of mechanisms to manipulate the mammalian host immune system and maintain homeostasis. However, the strategies employed by the microbiome to overcome host immune responses in invertebrates still remain to be understood. Here, we report that the gut microbiome in mosquitoes utilizes C-type lectins (mosGCTLs) to evade the bactericidal capacity of antimicrobial peptides (AMPs). Aedes aegypti mosGCTLs facilitate colonization by multiple bacterial strains. Furthermore, maintenance of the gut microbial flora relies on the expression of mosGCTLs in A. aegypti. Silencing the orthologues of mosGCTL in another major mosquito vector (Culex pipiens pallens) also impairs the survival of gut commensal bacteria. The gut microbiome stimulates the expression of mosGCTLs, which coat the bacterial surface and counteract AMP activity. Our study describes a mechanism by which the insect symbiotic microbiome offsets gut immunity to achieve homeostasis.
Collapse
|
58
|
Zhao W, Lu L, Yang P, Cui N, Kang L, Cui F. Organ-specific transcriptome response of the small brown planthopper toward rice stripe virus. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 70:60-72. [PMID: 26678499 DOI: 10.1016/j.ibmb.2015.11.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 11/15/2015] [Accepted: 11/30/2015] [Indexed: 05/13/2023]
Abstract
Rice stripe virus (RSV) causes rice stripe disease and is transmitted by the small brown planthopper (Laodelphax striatellus, SBPH) in a persistent, circulative, and propagative manner. The alimentary canal and salivary gland of SBPH play important roles in viral replication and transmission. However, little is known about the underlying molecular functions of these two organs in the interaction between RSV and SBPH. In this study, organ-specific transcriptomes of the alimentary canal and salivary gland were analyzed in viruliferous and naïve SBPH. The number of differentially expressed unigenes in the alimentary canal was considerably greater than that in the salivary gland after RSV infection, and only 23 unigenes were co-regulated in the two organs. In the alimentary canal, genes involved in lysosome, digestion and detoxification were activated upon RSV infection, whereas the genes related to DNA replication and repair were suppressed. RSV activated RNA transport and repressed the MAPK, mTOR, Wnt, and TGF-beta signaling pathways in the salivary gland. The overall immune reaction toward RSV was much stronger in the salivary gland than in the alimentary canal. RSV activated the pattern recognition molecules and Toll pathway in the salivary gland but inhibited these two reactions in the alimentary canal. The responses from reactive oxygen and the immune-responsive effectors were stronger in the salivary gland than in the alimentary canal after RSV infection. These findings provide clues on the roles of the two organs in confronting RSV infection and aid in the understanding of the interaction between RSV and SBPHs.
Collapse
Affiliation(s)
- Wan Zhao
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lixia Lu
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Pengcheng Yang
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, China
| | - Na Cui
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Le Kang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| | - Feng Cui
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
59
|
Smith RC, Vega-Rodríguez J, Jacobs-Lorena M. The Plasmodium bottleneck: malaria parasite losses in the mosquito vector. Mem Inst Oswaldo Cruz 2015. [PMID: 25185005 PMCID: PMC4156458 DOI: 10.1590/0074-0276130597] [Citation(s) in RCA: 153] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Nearly one million people are killed every year by the malaria parasite Plasmodium. Although the disease-causing forms of the parasite exist only in the human blood, mosquitoes of the genus Anopheles are the obligate vector for transmission. Here, we review the parasite life cycle in the vector and highlight the human and mosquito contributions that limit malaria parasite development in the mosquito host. We address parasite killing in its mosquito host and bottlenecks in parasite numbers that might guide intervention strategies to prevent transmission.
Collapse
Affiliation(s)
- Ryan C Smith
- Department of Molecular Microbiology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health and Immunology, Baltimore, MD, USA
| | - Joel Vega-Rodríguez
- Department of Molecular Microbiology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health and Immunology, Baltimore, MD, USA
| | - Marcelo Jacobs-Lorena
- Department of Molecular Microbiology, Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health and Immunology, Baltimore, MD, USA
| |
Collapse
|
60
|
Abstract
Small ubiquitin-like modifier (SUMO) modification modulates the expression of defense genes in Drosophila, activated by the Toll/nuclear factor-κB and immune-deficient/nuclear factor-κB signaling networks. We have, however, limited understanding of the SUMO-modulated regulation of the immune response and lack information on SUMO targets in the immune system. In this study, we measured the changes to the SUMO proteome in S2 cells in response to a lipopolysaccharide challenge and identified 1619 unique proteins in SUMO-enriched lysates. A confident set of 710 proteins represents the immune-induced SUMO proteome and analysis suggests that specific protein domains, cellular pathways, and protein complexes respond to immune stress. A small subset of the confident set was validated by in-bacto SUMOylation and shown to be bona-fide SUMO targets. These include components of immune signaling pathways such as Caspar, Jra, Kay, cdc42, p38b, 14-3-3ε, as well as cellular proteins with diverse functions, many being components of protein complexes, such as prosß4, Rps10b, SmD3, Tango7, and Aats-arg. Caspar, a human FAF1 ortholog that negatively regulates immune-deficient signaling, is SUMOylated at K551 and responds to treatment with lipopolysaccharide in cultured cells. Our study is one of the first to describe SUMO proteome for the Drosophila immune response. Our data and analysis provide a global framework for the understanding of SUMO modification in the host response to pathogens.
Collapse
|
61
|
Ma X, Li X, Dong S, Xia Q, Wang F. A Fas associated factor negatively regulates anti-bacterial immunity by promoting Relish degradation in Bombyx mori. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2015; 63:144-151. [PMID: 26101847 DOI: 10.1016/j.ibmb.2015.06.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/15/2015] [Accepted: 06/18/2015] [Indexed: 06/04/2023]
Abstract
Negative regulation is required to keep NF-κB-dependent immune response under tight control. In previous study, we have identified a Fas associated factor (FAF) family member in Bombyx mori, BmFAF, and proposed it may act as a negative regulator in immune response. In this study, we found knock-down of BmFAF by RNAi led to a remarkable increase in transcriptional level of several antimicrobial peptide genes, including BmCecropinA1 and BmMoricin, and higher survival rate to Gram-negative bacterial infection. We also confirmed the regulatory role of BmFAF in suppressing NF-κB-dependent transcription by employing an inducible promoter in BmE cells. Consistent with these physiological phenotypes, BmFAF suppressed the activity of the essential transcription factor, Relish, in IMD signaling pathway by promoting its proteasomal degradation through direct interaction. In addition, by constructing various truncation mutants, we further demonstrated that UBA domain in BmFAF is required for the inhibitory role, and potential ubiquitination also occurs in this domain. Taken together, our results suggest that BmFAF is a negative regulator of IMD pathway by mediating degradation of Relish.
Collapse
Affiliation(s)
- Xiaojuan Ma
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Xianyang Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Shifeng Dong
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Qingyou Xia
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China
| | - Fei Wang
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400716, China.
| |
Collapse
|
62
|
Molecular Analysis of a Recurrent Sarcoma Identifies a Mutation in FAF1. Sarcoma 2015; 2015:839182. [PMID: 25861239 PMCID: PMC4377510 DOI: 10.1155/2015/839182] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/11/2015] [Indexed: 11/25/2022] Open
Abstract
A patient presented with a recurrent sarcoma (diagnosed as leiomyosarcoma) 12 years after the removal of an initial cancer (diagnosed as extracompartmental osteosarcoma) distally on the same limb. Following surgery, the sarcoma and unaffected muscle and bone were subjected to measurements of DNA exome sequence, RNA and protein expression, and transcription factor binding. The investigation provided corroboration of the diagnosis leiomyosarcoma, as the major upregulations in this tumor comprise muscle-specific gene products and calcium-regulating molecules (calcium is an important second messenger in smooth muscle cells). A likely culprit for the disease is the point mutation S181G in FAF1, which may cause a loss of apoptotic function consecutive to transforming DNA damage. The RNA levels of genes for drug transport and metabolism were extensively skewed in the tumor tissue as compared to muscle and bone. The results suggest that the tumor represents a recurrence of a dormant metastasis from an originally misdiagnosed neoplasm. A loss of FAF1 function could cause constitutive WNT pathway activity (consistent with the downstream inductions of IGF2BP1 and E2F1 in this cancer). While the study has informed on drug transport and drug metabolism pharmacogenetics, it has fallen short of identifying a suitable target for molecular therapy.
Collapse
|
63
|
Visetnan S, Supungul P, Hirono I, Tassanakajon A, Rimphanitchayakit V. Activation of PmRelish from Penaeus monodon by yellow head virus. FISH & SHELLFISH IMMUNOLOGY 2015; 42:335-344. [PMID: 25463289 DOI: 10.1016/j.fsi.2014.11.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/12/2014] [Accepted: 11/13/2014] [Indexed: 06/04/2023]
Abstract
Humoral innate immune response against pathogenic infection is partly responsible by the Imd pathway in which a transcription factor Relish relays the infection signals to the nuclei for the expression of antimicrobial proteins. A PmRelish gene which encoded a protein of 1195 amino acids was cloned. The PmRelish was constitutively expressed in all tissues tested and mostly up-regulated upon YHV infection. In hemocytes, the PmRelish expression was up-regulated upon Vibrio harveyi, yellow head virus (YHV) and white spot syndrome virus (WSSV) challenges. Using dsRNA silencing of PmRelish gene, it was shown that the expression of penaeidin5 but not anti-lipopolysaccharide factor ALFPm3, crustinPm1 and penaeidin3 was under the regulation of Imd pathway. Under PmRelish silencing, the shrimp were more susceptible to infection by YHV with the 50% survival rate reduced from about 72 h to 42 h. The PmRelish was detected in the cytoplasm of all the hemocytes from both uninfected and YHV-infected shrimp. The accumulation of activated PmRelish in the nuclei was not clearly observed but the activated PmRelish was detected in the YHV-infected hemocytes by Western blot analysis. Thus, the PmRelish and, hence, the Imd pathway respond to the YHV infection.
Collapse
Affiliation(s)
- Suwattana Visetnan
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Road, Bangkok 10330, Thailand
| | - Premruethai Supungul
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Road, Bangkok 10330, Thailand; National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathum Thani 10120, Thailand
| | - Ikuo Hirono
- Laboratory of Genome Science, Tokyo University of Marine Science and Technology, Konan 4-5-7, Minato, Tokyo 108-8477, Japan
| | - Anchalee Tassanakajon
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Road, Bangkok 10330, Thailand
| | - Vichien Rimphanitchayakit
- Center of Excellence for Molecular Biology and Genomics of Shrimp, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Road, Bangkok 10330, Thailand.
| |
Collapse
|
64
|
Meyer SN, Amoyel M, Bergantiños C, de la Cova C, Schertel C, Basler K, Johnston LA. An ancient defense system eliminates unfit cells from developing tissues during cell competition. Science 2014; 346:1258236. [PMID: 25477468 DOI: 10.1126/science.1258236] [Citation(s) in RCA: 172] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Developing tissues that contain mutant or compromised cells present risks to animal health. Accordingly, the appearance of a population of suboptimal cells in a tissue elicits cellular interactions that prevent their contribution to the adult. Here we report that this quality control process, cell competition, uses specific components of the evolutionarily ancient and conserved innate immune system to eliminate Drosophila cells perceived as unfit. We find that Toll-related receptors (TRRs) and the cytokine Spätzle (Spz) lead to NFκB-dependent apoptosis. Diverse "loser" cells require different TRRs and NFκB factors and activate distinct pro-death genes, implying that the particular response is stipulated by the competitive context. Our findings demonstrate a functional repurposing of components of TRRs and NFκB signaling modules in the surveillance of cell fitness during development.
Collapse
Affiliation(s)
- S N Meyer
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - M Amoyel
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - C Bergantiños
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - C de la Cova
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - C Schertel
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - K Basler
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.
| | - L A Johnston
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
65
|
Mosquito immunity against arboviruses. Viruses 2014; 6:4479-504. [PMID: 25415198 PMCID: PMC4246235 DOI: 10.3390/v6114479] [Citation(s) in RCA: 130] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 10/30/2014] [Accepted: 11/11/2014] [Indexed: 01/03/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) pose a significant threat to global health, causing human disease with increasing geographic range and severity. The recent availability of the genome sequences of medically important mosquito species has kick-started investigations into the molecular basis of how mosquito vectors control arbovirus infection. Here, we discuss recent findings concerning the role of the mosquito immune system in antiviral defense, interactions between arboviruses and fundamental cellular processes such as apoptosis and autophagy, and arboviral suppression of mosquito defense mechanisms. This knowledge provides insights into co-evolutionary processes between vector and virus and also lays the groundwork for the development of novel arbovirus control strategies that target the mosquito vector.
Collapse
|
66
|
Azeez OI, Meintjes R, Chamunorwa JP. Fat body, fat pad and adipose tissues in invertebrates and vertebrates: the nexus. Lipids Health Dis 2014; 13:71. [PMID: 24758278 PMCID: PMC4005007 DOI: 10.1186/1476-511x-13-71] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 04/14/2014] [Indexed: 12/13/2022] Open
Abstract
The fat body in invertebrates was shown to participate in energy storage and homeostasis, apart from its other roles in immune mediation and protein synthesis to mention a few. Thus, sharing similar characteristics with the liver and adipose tissues in vertebrates. However, vertebrate adipose tissue or fat has been incriminated in the pathophysiology of metabolic disorders due to its role in production of pro-inflammatory cytokines. This has not been reported in the insect fat body. The link between the fat body and adipose tissue was examined in this review with the aim of determining the principal factors responsible for resistance to inflammation in the insect fat body. This could be the missing link in the prevention of metabolic disorders in vertebrates, occasioned by obesity.
Collapse
Affiliation(s)
- Odunayo Ibraheem Azeez
- Department of Anatomy and Physiology, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110 Pretoria, South Africa
- Department of Veterinary Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria
| | - Roy Meintjes
- Department of Anatomy and Physiology, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110 Pretoria, South Africa
| | - Joseph Panashe Chamunorwa
- Department of Anatomy and Physiology, Faculty of Veterinary Science, University of Pretoria, Private Bag X04, Onderstepoort 0110 Pretoria, South Africa
| |
Collapse
|
67
|
Myllymäki H, Valanne S, Rämet M. The Drosophila Imd Signaling Pathway. THE JOURNAL OF IMMUNOLOGY 2014; 192:3455-62. [DOI: 10.4049/jimmunol.1303309] [Citation(s) in RCA: 309] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
68
|
Ribeiro JMC, Genta FA, Sorgine MHF, Logullo R, Mesquita RD, Paiva-Silva GO, Majerowicz D, Medeiros M, Koerich L, Terra WR, Ferreira C, Pimentel AC, Bisch PM, Leite DC, Diniz MMP, Junior JLDSGV, Da Silva ML, Araujo RN, Gandara ACP, Brosson S, Salmon D, Bousbata S, González-Caballero N, Silber AM, Alves-Bezerra M, Gondim KC, Silva-Neto MAC, Atella GC, Araujo H, Dias FA, Polycarpo C, Vionette-Amaral RJ, Fampa P, Melo ACA, Tanaka AS, Balczun C, Oliveira JHM, Gonçalves RLS, Lazoski C, Rivera-Pomar R, Diambra L, Schaub GA, Garcia ES, Azambuja P, Braz GRC, Oliveira PL. An insight into the transcriptome of the digestive tract of the bloodsucking bug, Rhodnius prolixus. PLoS Negl Trop Dis 2014; 8:e2594. [PMID: 24416461 PMCID: PMC3886914 DOI: 10.1371/journal.pntd.0002594] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 11/04/2013] [Indexed: 12/14/2022] Open
Abstract
The bloodsucking hemipteran Rhodnius prolixus is a vector of Chagas' disease, which affects 7-8 million people today in Latin America. In contrast to other hematophagous insects, the triatomine gut is compartmentalized into three segments that perform different functions during blood digestion. Here we report analysis of transcriptomes for each of the segments using pyrosequencing technology. Comparison of transcript frequency in digestive libraries with a whole-body library was used to evaluate expression levels. All classes of digestive enzymes were highly expressed, with a predominance of cysteine and aspartic proteinases, the latter showing a significant expansion through gene duplication. Although no protein digestion is known to occur in the anterior midgut (AM), protease transcripts were found, suggesting secretion as pro-enzymes, being possibly activated in the posterior midgut (PM). As expected, genes related to cytoskeleton, protein synthesis apparatus, protein traffic, and secretion were abundantly transcribed. Despite the absence of a chitinous peritrophic membrane in hemipterans - which have instead a lipidic perimicrovillar membrane lining over midgut epithelia - several gut-specific peritrophin transcripts were found, suggesting that these proteins perform functions other than being a structural component of the peritrophic membrane. Among immunity-related transcripts, while lysozymes and lectins were the most highly expressed, several genes belonging to the Toll pathway - found at low levels in the gut of most insects - were identified, contrasting with a low abundance of transcripts from IMD and STAT pathways. Analysis of transcripts related to lipid metabolism indicates that lipids play multiple roles, being a major energy source, a substrate for perimicrovillar membrane formation, and a source for hydrocarbons possibly to produce the wax layer of the hindgut. Transcripts related to amino acid metabolism showed an unanticipated priority for degradation of tyrosine, phenylalanine, and tryptophan. Analysis of transcripts related to signaling pathways suggested a role for MAP kinases, GTPases, and LKBP1/AMP kinases related to control of cell shape and polarity, possibly in connection with regulation of cell survival, response of pathogens and nutrients. Together, our findings present a new view of the triatomine digestive apparatus and will help us understand trypanosome interaction and allow insights into hemipteran metabolic adaptations to a blood-based diet.
Collapse
Affiliation(s)
- José M. C. Ribeiro
- Section of Vector Biology, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Fernando A. Genta
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos H. F. Sorgine
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel Logullo
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael D. Mesquita
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabriela O. Paiva-Silva
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - David Majerowicz
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo Medeiros
- Instituto Nacional de Metrologia Qualidade e Tecnologia, Diretoria de Metrologia Aplicada às Ciências da Vida, Programa de Biotecnologia, Prédio 27, CEP 25250-020, Duque de Caxias, Rio de Janeiro, Brazil
| | - Leonardo Koerich
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, CEP 21944-970, Rio de Janeiro, Brazil
| | - Walter R. Terra
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Clélia Ferreira
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - André C. Pimentel
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo M. Bisch
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel C. Leite
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Michelle M. P. Diniz
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Lídio da S. G. V. Junior
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Center for Technological Innovation, Evandro Chagas Institute, Ananindeua, Pará, Brazil
| | - Manuela L. Da Silva
- Instituto Nacional de Metrologia Qualidade e Tecnologia, Diretoria de Metrologia Aplicada às Ciências da Vida, Programa de Biotecnologia, Prédio 27, CEP 25250-020, Duque de Caxias, Rio de Janeiro, Brazil
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo N. Araujo
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Parasitologia do Instituto de Ciências Biológicas da Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ana Caroline P. Gandara
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sébastien Brosson
- Institute for Molecular Biology and Medicine (IBMM), Université Libre de Bruxelles, Gosselies, Belgium
| | - Didier Salmon
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sabrina Bousbata
- Institute for Molecular Biology and Medicine (IBMM), Université Libre de Bruxelles, Gosselies, Belgium
| | | | - Ariel Mariano Silber
- Departamento de Parasitologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Michele Alves-Bezerra
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Katia C. Gondim
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mário Alberto C. Silva-Neto
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Georgia C. Atella
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Helena Araujo
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Institute for Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe A. Dias
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carla Polycarpo
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Raquel J. Vionette-Amaral
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Fampa
- Instituto de Biologia, DBA, UFRRJ, Seropédica, Rio de Janeiro, Brazil
| | - Ana Claudia A. Melo
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aparecida S. Tanaka
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Carsten Balczun
- Zoology/Parasitology Group, Ruhr-Universität, Bochum, Germany
| | - José Henrique M. Oliveira
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata L. S. Gonçalves
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cristiano Lazoski
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, CEP 21944-970, Rio de Janeiro, Brazil
| | - Rolando Rivera-Pomar
- Centro Regional de Estudios Genomicos, Universidad Nacional de La Plata, Florencio Varela, Argentina
- Centro de Bioinvestigaciones, Universidad Nacional del Noroeste de Buenos Aires, Pergamino, Argentina
| | - Luis Diambra
- Centro Regional de Estudios Genomicos, Universidad Nacional de La Plata, Florencio Varela, Argentina
| | | | - Elói S. Garcia
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Azambuja
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Glória R. C. Braz
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro L. Oliveira
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Bioquímica Médica, Programa de Biotecnologia e Biologia Molecular, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
69
|
Kleino A, Silverman N. The Drosophila IMD pathway in the activation of the humoral immune response. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2014; 42:25-35. [PMID: 23721820 PMCID: PMC3808521 DOI: 10.1016/j.dci.2013.05.014] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2013] [Revised: 05/17/2013] [Accepted: 05/17/2013] [Indexed: 05/08/2023]
Abstract
The IMD pathway signaling plays a pivotal role in the Drosophila defense against bacteria. During the last two decades, significant progress has been made in identifying the components and deciphering the molecular mechanisms underlying this pathway, including the means of bacterial sensing and signal transduction. While these findings have contributed to the understanding of the immune signaling in insects, they have also provided new insights in studying the mammalian NF-κB signaling pathways. Here, we summarize the current view of the IMD pathway focusing on how it regulates the humoral immune response of Drosophila.
Collapse
Affiliation(s)
- Anni Kleino
- Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | | |
Collapse
|
70
|
Manfredini F, Riba-Grognuz O, Wurm Y, Keller L, Shoemaker D, Grozinger CM. Sociogenomics of cooperation and conflict during colony founding in the fire ant Solenopsis invicta. PLoS Genet 2013; 9:e1003633. [PMID: 23950725 PMCID: PMC3738511 DOI: 10.1371/journal.pgen.1003633] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 05/30/2013] [Indexed: 11/18/2022] Open
Abstract
One of the fundamental questions in biology is how cooperative and altruistic behaviors evolved. The majority of studies seeking to identify the genes regulating these behaviors have been performed in systems where behavioral and physiological differences are relatively fixed, such as in the honey bee. During colony founding in the monogyne (one queen per colony) social form of the fire ant Solenopsis invicta, newly-mated queens may start new colonies either individually (haplometrosis) or in groups (pleometrosis). However, only one queen (the “winner”) in pleometrotic associations survives and takes the lead of the young colony while the others (the “losers”) are executed. Thus, colony founding in fire ants provides an excellent system in which to examine the genes underpinning cooperative behavior and how the social environment shapes the expression of these genes. We developed a new whole genome microarray platform for S. invicta to characterize the gene expression patterns associated with colony founding behavior. First, we compared haplometrotic queens, pleometrotic winners and pleometrotic losers. Second, we manipulated pleometrotic couples in order to switch or maintain the social ranks of the two cofoundresses. Haplometrotic and pleometrotic queens differed in the expression of genes involved in stress response, aging, immunity, reproduction and lipid biosynthesis. Smaller sets of genes were differentially expressed between winners and losers. In the second experiment, switching social rank had a much greater impact on gene expression patterns than the initial/final rank. Expression differences for several candidate genes involved in key biological processes were confirmed using qRT-PCR. Our findings indicate that, in S. invicta, social environment plays a major role in the determination of the patterns of gene expression, while the queen's physiological state is secondary. These results highlight the powerful influence of social environment on regulation of the genomic state, physiology and ultimately, social behavior of animals. The characterization of the genomic basis for complex behaviors is one of the major goals of biological research. The genomic state of an individual results from the interplay between its internal condition (the “nature”) and the external environment (the “nurture”), which may include the social environment. Colony founding in the fire ant Solenopsis invicta is a complex process that serves as a useful model for investigating how the interplay between genes and social environment shapes social behavior. Unrelated, newly mated S. invicta queens may start a new colony as a group, but ultimately only one queen will survive and gain full reproductive dominance. By uncovering the genetic basis for founding behavior in fire ants we therefore provide useful insights into how cooperative behavior evolved in a context that might be considered primitively eusocial, because newly mated queens in a founding association are morphologically, physiologically and genetically very similar and display no evident division of labor. Our results suggest that social environment (founding singly or in pairs, switching dominance rank vs. maintaining rank) is a much greater driver of gene expression changes than social rank itself, suggesting that social environment, and not reproductive state, is a key regulator of gene expression, physiology and ultimately, behavior.
Collapse
Affiliation(s)
- Fabio Manfredini
- Department of Entomology, Center for Pollinator Research, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA.
| | | | | | | | | | | |
Collapse
|
71
|
He Z, Wang P, Shi H, Si F, Hao Y, Chen B. Fas-associated factor 1 plays a negative regulatory role in the antibacterial immunity of Locusta migratoria. INSECT MOLECULAR BIOLOGY 2013; 22:389-398. [PMID: 23635314 DOI: 10.1111/imb.12029] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Insect immune responses are precisely regulated to maintain immune balance. In this study, the Fas-associated factor 1 (FAF1) gene of Locusta migratoria manilensis, a homologue of the caspar gene that functions as a specific negative regulator in the antibacterial immunity pathway, was cloned. Gene expression analysis showed that FAF1 was expressed throughout the developmental stages and in all tested tissues, but its transcription levels varied significantly. Thus, FAF1 appears to be tightly regulated and is probably involved in multiple physiological processes. In addition, the antimicrobial peptide gene prolixicin was cloned and characterized. After bacterial challenge, prolixicin was rapidly up-regulated, whereas FAF1 was markedly down-regulated. This result was consistent with the observation that prolixicin was hyperactivated when FAF1 was suppressed by RNA interference. Moreover, after bacterial infection, the survival rate of FAF1-knockdown locusts was much higher than that of the wild-type. Taken together, these findings strongly suggest that FAF1 shares a similar function as caspar in Drosophila and may be involved in the negative regulation of antibacterial immunity in locusts.
Collapse
Affiliation(s)
- Z He
- Institute of Entomology and Molecular Biology, College of Life Sciences, Chongqing Normal University, Chongqing, China
| | | | | | | | | | | |
Collapse
|
72
|
Shibata T, Sekihara S, Fujikawa T, Miyaji R, Maki K, Ishihara T, Koshiba T, Kawabata SI. Transglutaminase-catalyzed protein-protein cross-linking suppresses the activity of the NF-κB-like transcription factor relish. Sci Signal 2013; 6:ra61. [PMID: 23882120 DOI: 10.1126/scisignal.2003970] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cross-linking of proteins by mammalian transglutaminases (TGs) plays important roles in physiological phenomena such as blood coagulation and skin formation. We show that Drosophila TG suppressed innate immune signaling in the gut. RNA interference (RNAi) directed against TG reduced the life span of flies reared under conventional nonsterile conditions but not of those raised under germ-free conditions. In conventionally reared flies, TG RNAi enhanced the expression of genes encoding antimicrobial peptides in the immune deficiency (IMD) pathway. Wild-type flies that ingested gut lysates prepared from conventionally reared TG RNAi-treated flies had shorter life spans. In conventionally reared flies, TG RNAi triggered apoptosis in the gut and induced the nuclear translocation of Relish, the NF-κB (nuclear factor κB)-like transcription factor of the IMD pathway. Wild-type flies that ingested synthetic amine donors, which inhibit the TG-catalyzed protein-protein cross-linking reaction, showed nuclear translocation of Relish and enhanced expression of genes encoding IMD-controlled antimicrobial peptide genes in the gut. We conclude that TG-catalyzed Relish cross-linking suppressed the IMD signaling pathway to enable immune tolerance against commensal microbes.
Collapse
Affiliation(s)
- Toshio Shibata
- Department of Biology, Faculty of Sciences, Kyushu University, Fukuoka 812-8581, Japan
| | | | | | | | | | | | | | | |
Collapse
|
73
|
Kounatidis I, Ligoxygakis P. Drosophila as a model system to unravel the layers of innate immunity to infection. Open Biol 2013; 2:120075. [PMID: 22724070 PMCID: PMC3376734 DOI: 10.1098/rsob.120075] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Accepted: 04/25/2012] [Indexed: 12/11/2022] Open
Abstract
Innate immunity relies entirely upon germ-line encoded receptors, signalling components and effector molecules for the recognition and elimination of invading pathogens. The fruit fly Drosophila melanogaster with its powerful collection of genetic and genomic tools has been the model of choice to develop ideas about innate immunity and host–pathogen interactions. Here, we review current research in the field, encompassing all layers of defence from the role of the microbiota to systemic immune activation, and attempt to speculate on future directions and open questions.
Collapse
Affiliation(s)
- Ilias Kounatidis
- Laboratory of Genes and Development, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, UK
| | | |
Collapse
|
74
|
Kim CH, Muturi EJ. Effect of larval density and Sindbis virus infection on immune responses in Aedes aegypti. JOURNAL OF INSECT PHYSIOLOGY 2013; 59:604-610. [PMID: 23562781 DOI: 10.1016/j.jinsphys.2013.03.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 03/23/2013] [Accepted: 03/25/2013] [Indexed: 06/02/2023]
Abstract
Stressful environmental conditions during mosquito larval development may enhance susceptibility of adult mosquitoes to viral pathogens. Although anti-viral defense system in mosquitoes remains uncertain, stress-related enhancement of mosquito susceptibility to viral pathogens may be due to alteration of signaling pathways such as the Toll and immune deficiency (IMD) pathways. To test the influence of larval density and Sindbis virus (SINV) infection on mosquito Toll/Imd pathways, 100 or 200 Aedes aegypti larvae were reared in 2L of live oak (Quercus virginiana) leaf infusion and the adults were fed on SINV-infected (treatments) or non-infected (controls) bovine blood. SINV infection status and expression of genes encoding three antimicrobial peptides (cecropin, defensin, diptericin), an iron-binding protein (transferrin), and four regulators of Toll/Imd pathways (caspar, cactus, Rel1A, Rel2) were quantified by RT-qPCR at 7 and 14days post blood meals. Irrespective of larval density, females incubated for 14days after an infectious blood meal had significantly higher SINV titers compared to females from low density treatments that were incubated for 7days. For both larval densities and time intervals, there was significant down-regulation of the Toll/Imd regulator genes in SINV-infected mosquitoes compared to controls. At day 7 post-infection, there was significant down-regulation of cecropin, defensin, diptericin and transferrin in SINV-infected mosquitoes at low larval density but this effect was only observed for diptericin at high larval density. These genes remained suppressed on day 14, except cecropin which was significantly up-regulated at both larval densities, and transferrin which was similar to controls at low larval density. We conclude that SINV infection suppresses Toll/Imd pathways, but high larval density enables SINV to attain maximum titers in Ae. aegypti much earlier compared to low density treatments despite the up-regulation of cecropin.
Collapse
Affiliation(s)
- Chang-Hyun Kim
- Illinois Natural History Survey, University of Illinois, 1816 S. Oak St., Champaign, IL 61820, United States.
| | | |
Collapse
|
75
|
Aparicio R, Neyen C, Lemaitre B, Busturia A. dRYBP contributes to the negative regulation of the Drosophila Imd pathway. PLoS One 2013; 8:e62052. [PMID: 23596533 PMCID: PMC3626645 DOI: 10.1371/journal.pone.0062052] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 03/17/2013] [Indexed: 12/22/2022] Open
Abstract
The Drosophila humoral innate immune response fights infection by producing antimicrobial peptides (AMPs) through the microbe-specific activation of the Toll or the Imd signaling pathway. Upon systemic infection, the production of AMPs is both positively and negatively regulated to reach a balanced immune response required for survival. Here, we report the function of the dRYBP (drosophila Ring and YY1 Binding Protein) protein, which contains a ubiquitin-binding domain, in the Imd pathway. We have found that dRYBP contributes to the negative regulation of AMP production: upon systemic infection with Gram-negative bacteria, Diptericin expression is up-regulated in the absence of dRYBP and down-regulated in the presence of high levels of dRYBP. Epistatic analyses using gain and loss of function alleles of imd, Relish, or skpA and dRYBP suggest that dRYBP functions upstream or together with SKPA, a member of the SCF-E3-ubiquitin ligase complex, to repress the Imd signaling cascade. We propose that the role of dRYBP in the regulation of the Imd signaling pathway is to function as a ubiquitin adaptor protein together with SKPA to promote SCF-dependent proteasomal degradation of Relish. Beyond the identification of dRYBP as a novel component of Imd pathway regulation, our results also suggest that the evolutionarily conserved RYBP protein may be involved in the human innate immune response.
Collapse
Affiliation(s)
- Ricardo Aparicio
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Claudine Neyen
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Bruno Lemaitre
- Global Health Institute, School of Life Sciences, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Ana Busturia
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
76
|
Lee JJ, Park JK, Jeong J, Jeon H, Yoon JB, Kim EE, Lee KJ. Complex of Fas-associated factor 1 (FAF1) with valosin-containing protein (VCP)-Npl4-Ufd1 and polyubiquitinated proteins promotes endoplasmic reticulum-associated degradation (ERAD). J Biol Chem 2013; 288:6998-7011. [PMID: 23293021 DOI: 10.1074/jbc.m112.417576] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Fas-associated factor 1 (FAF1) is a ubiquitin receptor containing multiple ubiquitin-related domains including ubiquitin-associated (UBA), ubiquitin-like (UBL) 1, UBL2, and ubiquitin regulatory X (UBX). We previously showed that N-terminal UBA domain recognizes Lys(48)-ubiquitin linkage to recruit polyubiquitinated proteins and that a C-terminal UBX domain interacts with valosin-containing protein (VCP). This study shows that FAF1 interacts only with VCP complexed with Npl4-Ufd1 heterodimer, a requirement for the recruitment of polyubiquitinated proteins to UBA domain. Intriguingly, VCP association to C-terminal UBX domain regulates ubiquitin binding to N-terminal UBA domain without direct interaction between UBA and UBX domains. These interactions are well characterized by structural and biochemical analysis. VCP-Npl4-Ufd1 complex is known as the machinery required for endoplasmic reticulum-associated degradation. We demonstrate here that FAF1 binds to VCP-Npl4-Ufd1 complex via UBX domain and polyubiquitinated proteins via UBA domain to promote endoplasmic reticulum-associated degradation.
Collapse
Affiliation(s)
- Jae-Jin Lee
- Center for Cell Signaling and Drug Discovery Research, College of Pharmacy, Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Ewha Womans University, Seoul 120-750, Korea
| | | | | | | | | | | | | |
Collapse
|
77
|
Sebald J, Morettini S, Podhraski V, Lass-Flörl C, Lusser A. CHD1 contributes to intestinal resistance against infection by P. aeruginosa in Drosophila melanogaster. PLoS One 2012; 7:e43144. [PMID: 22912810 PMCID: PMC3418260 DOI: 10.1371/journal.pone.0043144] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 07/16/2012] [Indexed: 01/31/2023] Open
Abstract
Drosophila SNF2-type ATPase CHD1 catalyzes the assembly and remodeling of nucleosomal arrays in vitro and is involved in H3.3 incorporation in viin vivo during early embryo development. Evidence for a role as transcriptional regulator comes from its colocalization with elongating RNA polymerase II as well as from studies of fly Hsp70 transcription. Here we used microarray analysis to identify target genes of CHD1. We found a fraction of genes that were misregulated in Chd1 mutants to be functionally linked to Drosophila immune and stress response. Infection experiments using different microbial species revealed defects in host defense in Chd1-deficient adults upon oral infection with P. aeruginosa but not upon septic injury, suggesting a so far unrecognized role for CHD1 in intestinal immunity. Further molecular analysis showed that gut-specific transcription of antimicrobial peptide genes was overactivated in the absence of infection in Chd1 mutant flies. Moreover, microbial colonization of the intestine was elevated in Chd1 mutants and oral infection resulted in strong enrichment of bacteria in the body cavity indicating increased microbial passage across intestinal epithelia. However, we did not detect enhanced epithelial damage or alterations of the intestinal stem cell population. Collectively, our data provide evidence that intestinal resistance against infection by P. aeruginosa in Drosophila is linked to maintaining proper balance of gut-microbe interactions and that the chromatin remodeler CHD1 is involved in regulating this aspect.
Collapse
Affiliation(s)
- Johanna Sebald
- Division of Molecular Biology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Stefano Morettini
- Division of Molecular Biology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Valerie Podhraski
- Division of Molecular Biology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Cornelia Lass-Flörl
- Division of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Alexandra Lusser
- Division of Molecular Biology, Biocenter, Innsbruck Medical University, Innsbruck, Austria
- * E-mail:
| |
Collapse
|
78
|
Lee JJ, Kim YM, Jeong J, Bae DS, Lee KJ. Ubiquitin-associated (UBA) domain in human Fas associated factor 1 inhibits tumor formation by promoting Hsp70 degradation. PLoS One 2012; 7:e40361. [PMID: 22876279 PMCID: PMC3410879 DOI: 10.1371/journal.pone.0040361] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Accepted: 06/06/2012] [Indexed: 02/02/2023] Open
Abstract
Human Fas associated factor 1 (hFAF1) is a pro-apoptotic scaffolding protein containing ubiquitin-associating (UBA), ubiquitin like 1 and 2 (UBL1, UBL2), and ubiquitin regulatory X (UBX) domains. hFAF1 interacts with polyubiquitinated proteins via its N-terminal UBA domain and with valosin containing protein (VCP) via its C-terminal UBX domain. Overexpression of hFAF1 or its N-terminal UBA domain significantly increases cell death by increasing the degradation of polyubiquitinated proteins. In this study, we investigated whether hFAF1, whose expression level is reduced in cervical cancer, plays a role in tumor formation. We found that HeLa cells overexpressing full-length hFAF1 or the hFAF1 UBA domain alone, significantly suppressed the anchorage independent tumor growth in soft agar colony formation, increased cell death, and activated JNK and caspase 3. Employing UBA-specific tandem immunoprecipitation, we identified moieties specifically interacting with UBA domain of hFAF1, and found that polyubiquitinated Hsp70s are recruited to UBA domain. We also demonstrated that hFAF1 overexpression promotes Hsp70 degradation via the proteasome. We further found that mutating the UBA domain (I41N), as well as knocking down hFAF1 with specific RNAi, abolishs its ability to increase the proteasomal degradation of Hsp70. These findings suggest that hFAF1 inhibits tumor formation by increasing the degradation of Hsp70 mediated via its UBA domain.
Collapse
Affiliation(s)
- Jae-Jin Lee
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
| | - Young Mee Kim
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
| | - Jaeho Jeong
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
| | - Duk Soo Bae
- Department of Obstetrics and Gynecology, Samsung Medical Center, SungKyunKwan University, Seoul, Korea
| | - Kong-Joo Lee
- Division of Life and Pharmaceutical Sciences, Department of Bioinspired Science, Drug Discovery Research, College of Pharmacy, Center for Cell Signaling, Ewha Womans University, Seoul, Korea
- * E-mail:
| |
Collapse
|
79
|
Garver LS, Bahia AC, Das S, Souza-Neto JA, Shiao J, Dong Y, Dimopoulos G. Anopheles Imd pathway factors and effectors in infection intensity-dependent anti-Plasmodium action. PLoS Pathog 2012; 8:e1002737. [PMID: 22685401 PMCID: PMC3369948 DOI: 10.1371/journal.ppat.1002737] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 04/23/2012] [Indexed: 12/27/2022] Open
Abstract
The Anopheles gambiae immune response against Plasmodium falciparum, an etiological agent of human malaria, has been identified as a source of potential anti-Plasmodium genes and mechanisms to be exploited in efforts to control the malaria transmission cycle. One such mechanism is the Imd pathway, a conserved immune signaling pathway that has potent anti-P. falciparum activity. Silencing the expression of caspar, a negative regulator of the Imd pathway, or over-expressing rel2, an Imd pathway-controlled NFkappaB transcription factor, confers a resistant phenotype on A. gambiae mosquitoes that involves an array of immune effector genes. However, unexplored features of this powerful mechanism that may be essential for the implementation of a malaria control strategy still remain. Using RNA interference to singly or dually silence caspar and other components of the Imd pathway, we have identified genes participating in the anti-Plasmodium signaling module regulated by Caspar, each of which represents a potential target to achieve over-activation of the pathway. We also determined that the Imd pathway is most potent against the parasite's ookinete stage, yet also has reasonable activity against early oocysts and lesser activity against late oocysts. We further demonstrated that caspar silencing alone is sufficient to induce a robust anti-P. falciparum response even in the relative absence of resident gut microbiota. Finally, we established the relevance of the Imd pathway components and regulated effectors TEP1, APL1, and LRIM1 in parasite infection intensity-dependent defense, thereby shedding light on the relevance of laboratory versus natural infection intensity models. Our results highlight the physiological considerations that are integral to a thoughtful implementation of Imd pathway manipulation in A. gambiae as part of an effort to limit the malaria transmission cycle, and they reveal a variety of previously unrecognized nuances in the Imd-directed immune response against P. falciparum. The immune response of the mosquito vector of Plasmodium has proven to possess powerful anti-Plasmodium defense capabilities. As the major regulators of these immune responses, signaling pathways, particularly the Imd pathway which seems especially capable of eliminating malaria parasites, have become attractive candidates targets for malaria-control interventions. Although the general anti-parasitic activity of the Imd pathway has been established, the particular components of the pathway involved and the physiological conditions under which the pathway is capable of limiting infection are mostly unknown. Awareness of these major players and conditions is crucial for adapting the Imd pathway into an intervention strategy. We report that while several members of the Imd pathway are critical for such a response, others are dispensable. We also show that timing of the response with regard to infection and intensity of infection exposure both influence the effectiveness of an Imd-derived anti-Plasmodium response while the status of the gut flora does not. Taken together, this data lays the essential groundwork for effective intervention based on manipulation of this pathway that can severely limit mosquito infection with human malaria parasites.
Collapse
Affiliation(s)
- Lindsey S. Garver
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Ana C. Bahia
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Suchismita Das
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jayme A. Souza-Neto
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Jessica Shiao
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
80
|
Polydnavirus Ank proteins bind NF-κB homodimers and inhibit processing of Relish. PLoS Pathog 2012; 8:e1002722. [PMID: 22654665 PMCID: PMC3359993 DOI: 10.1371/journal.ppat.1002722] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Accepted: 04/12/2012] [Indexed: 12/25/2022] Open
Abstract
Recent studies have greatly increased understanding of how the immune system of insects responds to infection, whereas much less is known about how pathogens subvert immune defenses. Key regulators of the insect immune system are Rel proteins that form Nuclear Factor-κB (NF-κB) transcription factors, and inhibitor κB (IκB) proteins that complex with and regulate NF-κBs. Major mortality agents of insects are parasitoid wasps that carry immunosuppressive polydnaviruses (PDVs). Most PDVs encode ank genes that share features with IκBs, while our own prior studies suggested that two ank family members from Microplitis demolitor bracovirus (MdBV) (Ank-H4 and Ank-N5) behave as IκB mimics. However, the binding affinities of these viral mimics for Rel proteins relative to endogenous IκBs remained unclear. Surface plasmon resonance (SPR) and co-immunoprecipitation assays showed that the IκB Cactus from Drosophila bound Dif and Dorsal homodimers more strongly than Relish homodimers. Ank-H4 and –N5 bound Dif, Dorsal and Relish homodimers with higher affinity than the IκB domain of Relish (Rel-49), and also bound Relish homodimers more strongly than Cactus. Ank-H4 and –N5 inhibited processing of compound Relish and reduced the expression of several antimicrobial peptide genes regulated by the Imd signaling pathway in Drosophila mbn2 cells. Studies conducted in the natural host Pseudoplusia includens suggested that parasitism by M. demolitor also activates NF-κB signaling and that MdBV inhibits this response. Overall, our data provide the first quantitative measures of insect and viral IκB binding affinities, while also showing that viral mimics disable Relish processing. Central to the study of host-pathogen interactions is understanding how the immune system of hosts responds to infection, and reciprocally how pathogens subvert host defenses. In the case of insects, understanding of how the immune system responds to infection greatly exceeds understanding of pathogen counterstrategies. Parasitoid wasps are key mortality agents of insects. Thousands of wasp species have also evolved a symbiotic relationship with large DNA viruses in the family Polydnaviridae whose primary function is to deliver immunosuppressive virulence genes to the insect hosts that wasps parasitize. The function of most PDV-encoded virulence genes, however, remains unknown. In this article, we investigated the function of two ank gene family members from Microplitis demolitor bracovirus (MdBV). Our results indicate that Ank-H4 and Ank-N5 function as mimics of IκB proteins, which regulate a family of transcription factors called NF-κBs that control many genes of the insect immune system. IκBs and NF-κBs also function as key regulators of the mammalian immune system. Our results thus suggest that viral Ank proteins subvert the immune system of host insects by targeting conserved signaling pathways used by a diversity of organisms.
Collapse
|
81
|
Valanne S, Kallio J, Kleino A, Rämet M. Large-scale RNAi screens add both clarity and complexity to Drosophila NF-κB signaling. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 37:9-18. [PMID: 21930155 DOI: 10.1016/j.dci.2011.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Revised: 09/02/2011] [Accepted: 09/03/2011] [Indexed: 05/31/2023]
Abstract
NF-κB signaling is an immune response mechanism remarkably conserved through phylogeny. The genetically tractable model animal Drosophila melanogaster is an important model organism for studying NF-κB signaling in the immune response. Fruit flies have two NF-κB signaling pathways: the Toll and the Imd pathway. Traditional genetic screens have revealed many important aspects about the regulation of Drosophila NF-κB signaling and have helped us to also understand the immune response in humans. For example, the discovery that Toll like receptors are the main immune signaling molecules in mammals was based on work in flies. During the past decade high throughput RNA interference (RNAi)-based screening in cultured Drosophila cells has become a common method for identifying novel genes required for numerous cellular processes including NF-κB signaling. These screens have identified many novel positive and negative regulators of Drosophila NF-κB signaling thus enhancing our understanding of these signaling cascades.
Collapse
Affiliation(s)
- Susanna Valanne
- Laboratory of Experimental Immunology, Institute of Biomedical Technology, University of Tampere, 33520 Tampere, Finland
| | | | | | | |
Collapse
|
82
|
Choi IK, Hyun S. Conserved microRNA miR-8 in fat body regulates innate immune homeostasis in Drosophila. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2012; 37:50-54. [PMID: 22210547 DOI: 10.1016/j.dci.2011.12.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 12/13/2011] [Accepted: 12/14/2011] [Indexed: 05/31/2023]
Abstract
Antimicrobial peptides (AMPs) constitute a major arm of the innate immune system across diverse organisms. In Drosophila, septic injury by microbial pathogens rapidly induces the production of the AMPs in fat body via well elucidated pathways such as Toll and IMD. However, several epithelial tissues were reported to locally express AMPs without septic injury via poorly characterized ways. Here, we report that microRNA miR-8 regulates the levels of AMPs basally expressed in Drosophila. The levels of AMPs such as Drosomycin and Diptericin are significantly increased in miR-8 null animals in non-pathogen stimulated conditions. Analysis of various larval tissues revealed that the increase of Drosomycin is fat body specific. Supporting this observation, re-introduction of miR-8 only in the fat body restored the altered AMP expression in miR-8 null flies. Although loss of miR-8 impedes PI3K in the fat body, inhibition of PI3K does not phenocopy the AMP expression of miR-8 null flies, indicating that miR-8 regulates AMP independently of PI3K. Together, our findings suggest a role of miR-8 in systemic immune homeostasis in generally non-pathogenic conditions in flies.
Collapse
Affiliation(s)
- In Kyou Choi
- School of Biological Sciences, Chung-Ang University, Seoul 156-756, Republic of Korea
| | | |
Collapse
|
83
|
A shared role for RBF1 and dCAP-D3 in the regulation of transcription with consequences for innate immunity. PLoS Genet 2012; 8:e1002618. [PMID: 22496667 PMCID: PMC3320600 DOI: 10.1371/journal.pgen.1002618] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 02/08/2012] [Indexed: 12/27/2022] Open
Abstract
Previously, we discovered a conserved interaction between RB proteins and the Condensin II protein CAP-D3 that is important for ensuring uniform chromatin condensation during mitotic prophase. The Drosophila melanogaster homologs RBF1 and dCAP-D3 co-localize on non-dividing polytene chromatin, suggesting the existence of a shared, non-mitotic role for these two proteins. Here, we show that the absence of RBF1 and dCAP-D3 alters the expression of many of the same genes in larvae and adult flies. Strikingly, most of the genes affected by the loss of RBF1 and dCAP-D3 are not classic cell cycle genes but are developmentally regulated genes with tissue-specific functions and these genes tend to be located in gene clusters. Our data reveal that RBF1 and dCAP-D3 are needed in fat body cells to activate transcription of clusters of antimicrobial peptide (AMP) genes. AMPs are important for innate immunity, and loss of either dCAP-D3 or RBF1 regulation results in a decrease in the ability to clear bacteria. Interestingly, in the adult fat body, RBF1 and dCAP-D3 bind to regions flanking an AMP gene cluster both prior to and following bacterial infection. These results describe a novel, non-mitotic role for the RBF1 and dCAP-D3 proteins in activation of the Drosophila immune system and suggest dCAP-D3 has an important role at specific subsets of RBF1-dependent genes. The retinoblastoma protein (pRB) is a tumor suppressor protein known for its ability to repress transcription of E2F-dependent genes and induce cell cycle arrest. We have previously shown that RB proteins in Drosophila and human cells interact with the Condensin II subunit, CAP-D3, in an E2F-independent manner. Condensins promote condensation of chomosomes in mitosis. Our previous studies suggested that the Drosophila pRB and CAP-D3 homologs, RBF1 and dCAP-D3, co-localize on DNA and may share a function in cells that never undergo mitosis. In this study, we show that one non-mitotic function shared between RBF1 and dCAP-D3 is the regulation of many non-cell-cycle-related, clustered, and cell-type-specific transcripts including a conserved family of genes that are important for the immune response in the fly. In fact, results show that normal levels of dCAP-D3 and RBF1 expression are necessary for the ability of the fly to clear infection with human bacterial pathogens. This work demonstrates that dCAP-D3 proteins can regulate a unique subset of RBF1-dependent transcripts in vivo and identifies a novel role for both RBF1 and dCAP-D3 protein in activation of innate immune genes, which may be conserved in human cells.
Collapse
|
84
|
Telleria EL, Sant'Anna MRV, Ortigão-Farias JR, Pitaluga AN, Dillon VM, Bates PA, Traub-Csekö YM, Dillon RJ. Caspar-like gene depletion reduces Leishmania infection in sand fly host Lutzomyia longipalpis. J Biol Chem 2012; 287:12985-93. [PMID: 22375009 DOI: 10.1074/jbc.m111.331561] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Female phlebotomine sand flies Lutzomyia longipalpis naturally harbor populations of the medically important Leishmania infantum (syn. Leishmania chagasi) parasite in the gut, but the extent to which the parasite interacts with the immune system of the insect vector is unknown. To investigate the sand fly immune response and its interaction with the Leishmania parasite, we identified a homologue for caspar, a negative regulator of immune deficiency signaling pathway. We found that feeding antibiotics to adult female L. longipalpis resulted in an up-regulation of caspar expression relative to controls. caspar was differentially expressed when females were fed on gram-negative and gram-positive bacterial species. caspar expression was significantly down-regulated in females between 3 and 6 days after a blood feed containing Leishmania mexicana amastigotes. RNA interference was used to deplete caspar expression in female L. longipalpis, which were subsequently fed with Leishmania in a blood meal. Sand fly gut populations of both L. mexicana and L. infantum were significantly reduced in caspar-depleted females. The prevalence of L. infantum infection in the females fell from 85 to 45%. Our results provide the first insight into the operation of immune homeostasis in phlebotomine sand flies during the growth of bacterial and Leishmania populations in the digestive tract. We have demonstrated that the activation of the sand fly immune system, via depletion of a single gene, can lead to the abortion of Leishmania development and the disruption of transmission by the phlebotomine sand fly.
Collapse
Affiliation(s)
- Erich L Telleria
- Instituto Oswaldo Cruz, Fiocruz, Av. Brasil 4365, 21045-900, Rio de Janeiro, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
85
|
Abstract
The deleterious and sometimes fatal outcomes of bacterial infectious diseases are the net result of the interactions between the pathogen and the host, and the genetically tractable fruit fly, Drosophila melanogaster, has emerged as a valuable tool for modeling the pathogen-host interactions of a wide variety of bacteria. These studies have revealed that there is a remarkable conservation of bacterial pathogenesis and host defence mechanisms between higher host organisms and Drosophila. This review presents an in-depth discussion of the Drosophila immune response, the Drosophila killing model, and the use of the model to examine bacterial-host interactions. The recent introduction of the Drosophila model into the oral microbiology field is discussed, specifically the use of the model to examine Porphyromonas gingivalis-host interactions, and finally the potential uses of this powerful model system to further elucidate oral bacterial-host interactions are addressed.
Collapse
Affiliation(s)
- Christina O Igboin
- Division of Oral Biology, College of Dentistry, The Ohio State University, Columbus, Ohio, USA
| | | | | |
Collapse
|
86
|
Bonizzoni M, Dunn WA, Campbell CL, Olson KE, Marinotti O, James AA. Strain Variation in the Transcriptome of the Dengue Fever Vector, Aedes aegypti. G3 (BETHESDA, MD.) 2012; 2:103-14. [PMID: 22384387 PMCID: PMC3276191 DOI: 10.1534/g3.111.001107] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 11/15/2011] [Indexed: 11/18/2022]
Abstract
Studies of transcriptome dynamics provide a basis for understanding functional elements of the genome and the complexity of gene regulation. The dengue vector mosquito, Aedes aegypti, exhibits great adaptability to diverse ecological conditions, is phenotypically polymorphic, and shows variation in vectorial capacity to arboviruses. Previous genome sequencing showed richness in repetitive DNA and transposable elements that can contribute to genome plasticity. Population genetic studies revealed a varying degree of worldwide genetic polymorphism. However, the extent of functional genetic polymorphism across strains is unknown. The transcriptomes of three Ae. aegypti strains, Chetumal (CTM), Rexville D-Puerto Rico (Rex-D) and Liverpool (LVP), were compared. CTM is more susceptible than Rex- D to infection by dengue virus serotype 2. A total of 4188 transcripts exhibit either no or small variation (<2-fold) among sugar-fed samples of the three strains and between sugar- and blood-fed samples within each strain, corresponding most likely to genes encoding products necessary for vital functions. Transcripts enriched in blood-fed mosquitoes encode proteins associated with catalytic activities, molecular transport, metabolism of lipids, carbohydrates and amino acids, and functions related to blood digestion and the progression of the gonotropic cycle. Significant qualitative and quantitative differences were found in individual transcripts among strains including differential representation of paralogous gene products. The majority of immunity-associated transcripts decreased in accumulation after a bloodmeal and the results are discussed in relation to the different susceptibility of CTM and Rex-D mosquitoes to DENV2 infection.
Collapse
Affiliation(s)
| | - W. Augustine Dunn
- Department of Molecular Biology and Biochemistry, and
- Institute for Genomics and Bioinformatics, University of California, Irvine, California 92697
| | | | - Ken E. Olson
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado 80523
| | | | - Anthony A. James
- Department of Molecular Biology and Biochemistry, and
- Department of Microbiology and Molecular Genetics, University of California, California, Irvine 92697
| |
Collapse
|
87
|
Zou Z, Souza-Neto J, Xi Z, Kokoza V, Shin SW, Dimopoulos G, Raikhel A. Transcriptome analysis of Aedes aegypti transgenic mosquitoes with altered immunity. PLoS Pathog 2011; 7:e1002394. [PMID: 22114564 PMCID: PMC3219725 DOI: 10.1371/journal.ppat.1002394] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2011] [Accepted: 10/07/2011] [Indexed: 12/17/2022] Open
Abstract
The mosquito immune system is involved in pathogen-elicited defense responses. The NF-κB factors REL1 and REL2 are downstream transcription activators of Toll and IMD immune pathways, respectively. We have used genome-wide microarray analyses to characterize fat-body-specific gene transcript repertoires activated by either REL1 or REL2 in two transgenic strains of the mosquito Aedes aegypti. Vitellogenin gene promoter was used in each transgenic strain to ectopically express either REL1 (REL1+) or REL2 (REL2+) in a sex, tissue, and stage specific manner. There was a significant change in the transcript abundance of 297 (79 up- and 218 down-regulated) and 299 (123 up- and 176 down-regulated) genes in fat bodies of REL1+ and REL2+, respectively. Over half of the induced genes had predicted functions in immunity, and a large group of these was co-regulated by REL1 and REL2. By generating a hybrid transgenic strain, which ectopically expresses both REL1 and REL2, we have shown a synergistic action of these NF-κB factors in activating immune genes. The REL1+ immune transcriptome showed a significant overlap with that of cactus (RNAi)-depleted mosquitoes (50%). In contrast, the REL2+ -regulated transcriptome differed from the relatively small group of gene transcripts regulated by RNAi depletion of a putative inhibitor of the IMD pathway, caspar (35 up- and 140 down-regulated), suggesting that caspar contributes to regulation of a subset of IMD-pathway controlled genes. Infections of the wild type Ae. aegypti with Plasmodium gallinaceum elicited the transcription of a distinct subset of immune genes (76 up- and 25 down-regulated) relative to that observed in REL1+ and REL2+ mosquitoes. Considerable overlap was observed between the fat body transcriptome of Plasmodium-infected mosquitoes and that of mosquitoes with transiently depleted PIAS, an inhibitor of the JAK-STAT pathway. PIAS gene silencing reduced Plasmodium proliferation in Ae. aegypti, indicating the involvement of the JAK-STAT pathway in anti-Plasmodium defense in this infection model.
Collapse
Affiliation(s)
- Zhen Zou
- Department of Entomology and the Institute for Integrative Genome Biology, University of California, Riverside, California, United States of America
| | - Jayme Souza-Neto
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Zhiyong Xi
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Vladimir Kokoza
- Department of Entomology and the Institute for Integrative Genome Biology, University of California, Riverside, California, United States of America
| | - Sang Woon Shin
- Department of Entomology and the Institute for Integrative Genome Biology, University of California, Riverside, California, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Alexander Raikhel
- Department of Entomology and the Institute for Integrative Genome Biology, University of California, Riverside, California, United States of America
| |
Collapse
|
88
|
Valanne S, Wang JH, Rämet M. The Drosophila Toll signaling pathway. THE JOURNAL OF IMMUNOLOGY 2011; 186:649-56. [PMID: 21209287 DOI: 10.4049/jimmunol.1002302] [Citation(s) in RCA: 633] [Impact Index Per Article: 45.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The identification of the Drosophila melanogaster Toll pathway cascade and the subsequent characterization of TLRs have reshaped our understanding of the immune system. Ever since, Drosophila NF-κB signaling has been actively studied. In flies, the Toll receptors are essential for embryonic development and immunity. In total, nine Toll receptors are encoded in the Drosophila genome, including the Toll pathway receptor Toll. The induction of the Toll pathway by gram-positive bacteria or fungi leads to the activation of cellular immunity as well as the systemic production of certain antimicrobial peptides. The Toll receptor is activated when the proteolytically cleaved ligand Spatzle binds to the receptor, eventually leading to the activation of the NF-κB factors Dorsal-related immunity factor or Dorsal. In this study, we review the current literature on the Toll pathway and compare the Drosophila and mammalian NF-κB pathways.
Collapse
Affiliation(s)
- Susanna Valanne
- Laboratory of Experimental Immunology, Institute of Medical Technology, University of Tampere, 33014 Tampere, Finland
| | | | | |
Collapse
|
89
|
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
|
90
|
Radyuk SN, Michalak K, Klichko VI, Benes J, Orr WC. Peroxiredoxin 5 modulates immune response in Drosophila. Biochim Biophys Acta Gen Subj 2010; 1800:1153-63. [PMID: 20600624 DOI: 10.1016/j.bbagen.2010.06.010] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2010] [Revised: 06/16/2010] [Accepted: 06/21/2010] [Indexed: 10/19/2022]
Abstract
BACKGROUND Peroxiredoxins are redox-sensing enzymes with multiple cellular functions. Previously, we reported on the potent antioxidant function of Drosophila peroxiredoxin 5 (dPrx5). Studies with mammalian and human cells suggest that peroxiredoxins can modulate immune-related signaling. METHODS Survivorship studies and bacteriological analysis were used to determine resistance of flies to fungal and bacterial infections. RT-PCR and immunoblot analyses determined expression of dPrx5 and immunity factors in response to bacterial challenge. Double mutants for dprx5 gene and genes comprising the Imd/Relish and dTak1/Basket branches of the immune signaling pathways were used in epistatic analysis. RESULTS The dprx5 mutant flies were more resistant to bacterial infection than controls, while flies overexpressing dPrx5 were more susceptible. The enhanced resistance to bacteria was accompanied by rapid induction of the Imd-dependent antimicrobial peptides, phosphorylation of the JNK kinase Basket and altered transcriptional profiling of the transient response genes, puckered, ets21C and relish, while the opposite effects were observed in flies over-expressing dPrx5. Epistatic analysis of double mutants, using attacin D and Puckered as read outs of activation of the Imd and JNK pathways, implicated dPrx5 function in the control of the dTak1-JNK arm of immune signaling. CONCLUSIONS Differential effects on fly survivorship suggested a trade-off between the antioxidant and immune functions of dPrx5. Molecular and epistatic analyses identified dPrx5 as a negative regulator in the dTak1-JNK arm of immune signaling. GENERAL SIGNIFICANCE Our findings suggest that peroxiredoxins play an important modulatory role in the Drosophila immune response.
Collapse
Affiliation(s)
- Svetlana N Radyuk
- Department of Biological Sciences, Southern Methodist University, 6501 Airline Rd., Room 113, Dallas, TX 75275, USA.
| | | | | | | | | |
Collapse
|
91
|
Cirimotich CM, Dong Y, Garver LS, Sim S, Dimopoulos G. Mosquito immune defenses against Plasmodium infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2010; 34:387-95. [PMID: 20026176 PMCID: PMC3462653 DOI: 10.1016/j.dci.2009.12.005] [Citation(s) in RCA: 152] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2009] [Revised: 12/07/2009] [Accepted: 12/08/2009] [Indexed: 05/11/2023]
Abstract
The causative agent of malaria, Plasmodium, has to undergo complex developmental transitions and survive attacks from the mosquito's innate immune system to achieve transmission from one host to another through the vector. Here we discuss recent findings on the role of the mosquito's innate immune signaling pathways in preventing infection by the Plasmodium parasite, the identification and mechanistic description of novel anti-parasite molecules, the role that natural bacteria harbored in the mosquito midgut might play in this immune defense and the crucial parasite and vector molecules that mediate midgut infection.
Collapse
Affiliation(s)
- Chris M Cirimotich
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe St., Baltimore, MD 21205, USA
| | | | | | | | | |
Collapse
|
92
|
Song J, Park JK, Lee JJ, Choi YS, Ryu KS, Kim JH, Kim E, Lee KJ, Jeon YH, Kim EE. Structure and interaction of ubiquitin-associated domain of human Fas-associated factor 1. Protein Sci 2010; 18:2265-76. [PMID: 19722279 DOI: 10.1002/pro.237] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Fas-associated factor (FAF)-1 is a multidomain protein that was first identified as a member of the Fas death-inducing signaling complex, but later found to be involved in various biological processes. Although the exact mechanisms are not clear, FAF1 seems to play an important role in cancer, asbestos-induced mesotheliomas, and Parkinson's disease. It interacts with polyubiquitinated proteins, Hsp70, and p97/VCP (valosin-containing protein), in addition to the proteins of the Fas-signaling pathway. We have determined the crystal structure of the ubiquitin-associated domain of human FAF1 (hFAF1-UBA) and examined its interaction with ubiquitin and ubiquitin-like proteins using nuclear magnetic resonance. hFAF1-UBA revealed a canonical three-helical bundle that selectively binds to mono- and di-ubiquitin (Lys48-linked), but not to SUMO-1 (small ubiquitin-related modifier 1) or NEDD8 (neural precursor cell expressed, developmentally down-regulated 8). The interaction between hFAF1-UBA and di-ubiquitin involves hydrophobic interaction accompanied by a transition in the di-ubiquitin conformation. These results provide structural insight into the mechanism of polyubiquitin recognition by hFAF1-UBA.
Collapse
Affiliation(s)
- Jinsue Song
- Magnetic Resonance Team, Korea Basic Science Institute, 804-1 Yangchung-Ri, Ochang, Chungbuk, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Hetru C, Hoffmann JA. NF-kappaB in the immune response of Drosophila. Cold Spring Harb Perspect Biol 2009; 1:a000232. [PMID: 20457557 DOI: 10.1101/cshperspect.a000232] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The nuclear factor kappaB (NF-kappaB) pathways play a major role in Drosophila host defense. Two recognition and signaling cascades control this immune response. The Toll pathway is activated by Gram-positive bacteria and by fungi, whereas the immune deficiency (Imd) pathway responds to Gram-negative bacterial infection. The basic mechanisms of recognition of these various types of microbial infections by the adult fly are now globally understood. Even though some elements are missing in the intracellular pathways, numerous proteins and interactions have been identified. In this article, we present a general picture of the immune functions of NF-kappaB in Drosophila with all the partners involved in recognition and in the signaling cascades.
Collapse
Affiliation(s)
- Charles Hetru
- Centre National de la Recherche Scientifique, Institute of Molecular and Cellular Biology, 15 rue René Descartes, 67084 Strasbourg, France
| | | |
Collapse
|
94
|
Wei Y, Xiao Q, Zhang T, Mou Z, You J, Ma WJ. Differential regulation of mRNA stability controls the transient expression of genes encoding Drosophila antimicrobial peptide with distinct immune response characteristics. Nucleic Acids Res 2009; 37:6550-61. [PMID: 19726583 PMCID: PMC2770663 DOI: 10.1093/nar/gkp693] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The tight regulation of transiently expressed antimicrobial peptides (AMPs) with a distinct antimicrobial spectrum and different expression kinetics contributes greatly to the properly regulated immune response for resistance to pathogens and for the maintenance of mutualistic microbiota in Drosophila. The important role of differential regulation of AMP expression at the posttranscriptional level needs to be elucidated. It was observed that the highly expressed Cecropin A1 (CecA1) mRNA encoding a broad antimicrobial spectrum AMP against both bacteria and fungi decayed more quickly than did the moderately expressed Diptericin mRNA encoding AMP against Gram negative bacteria. The mRNA stability of AMPs is differentially regulated and is attributed to the specific interaction between cis-acting ARE in 3'-UTR of AMP mRNA and the RNA destabilizing protein transactor Tis11 as shown in co-immunoprecipitation of the Tis11 RNP complex with CecA1 mRNA but not other AMP mRNA. The p38MAPK was further demonstrated to play a crucial role in stabilizing ARE-bearing mRNAs by inhibiting Tis11-mediated degradation in LPS induced AMP expression. This evidence suggests an evolutionarily conserved and functionally important molecular basis for and effective approach to exact control of AMP gene expression. These mechanisms thereby orchestrate a well balanced and dynamic antimicrobial spectrum of innate immunity to resist infection and maintain resident microbiota properly.
Collapse
Affiliation(s)
- Youheng Wei
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences & Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | | | |
Collapse
|
95
|
Garver LS, Dong Y, Dimopoulos G. Caspar controls resistance to Plasmodium falciparum in diverse anopheline species. PLoS Pathog 2009; 5:e1000335. [PMID: 19282971 PMCID: PMC2647737 DOI: 10.1371/journal.ppat.1000335] [Citation(s) in RCA: 186] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2008] [Accepted: 02/12/2009] [Indexed: 02/04/2023] Open
Abstract
Immune responses mounted by the malaria vector Anopheles gambiae are largely regulated by the Toll and Imd (immune deficiency) pathways via the NF-kappaB transcription factors Rel1 and Rel2, which are controlled by the negative regulators Cactus and Caspar, respectively. Rel1- and Rel2-dependent transcription in A. gambiae has been shown to be particularly critical to the mosquito's ability to manage infection with the rodent malaria parasite Plasmodium berghei. Using RNA interference to deplete the negative regulators of these pathways, we found that Rel2 controls resistance of A. gambiae to the human malaria parasite Plasmodium falciparum, whereas Rel 1 activation reduced infection levels. The universal relevance of this defense system across Anopheles species was established by showing that caspar silencing also prevents the development of P. falciparum in the major malaria vectors of Asia and South America, A. stephensi and A. albimanus, respectively. Parallel studies suggest that while Imd pathway activation is most effective against P. falciparum, the Toll pathway is most efficient against P. berghei, highlighting a significant discrepancy between the human pathogen and its rodent model. High throughput gene expression analyses identified a plethora of genes regulated by the activation of the two Rel factors and revealed that the Toll pathway played a more diverse role in mosquito biology than the Imd pathway, which was more immunity-specific. Further analyses of key anti-Plasmodium factors suggest they may be responsible for the Imd pathway-mediated resistance phenotype. Additionally, we found that the fitness cost caused by Rel2 activation through caspar gene silencing was undetectable in sugar-fed, blood-fed, and P. falciparum-infected female A. gambiae, while activation of the Toll pathway's Rel1 had a major impact. This study describes for the first time a single gene that influences an immune mechanism that is able to abort development of P. falciparum in Anopheline species. Further, this study addresses aspects of the molecular, evolutionary, and physiological consequences of the observed phenotype. These findings have implications for malaria control since broad-spectrum immune activation in diverse anopheline species offers a viable and strategic approach to develop novel malaria control methods worldwide.
Collapse
Affiliation(s)
- Lindsey S. Garver
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Yuemei Dong
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
96
|
Guntermann S, Primrose DA, Foley E. Dnr1-dependent regulation of the Drosophila immune deficiency signaling pathway. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2009; 33:127-134. [PMID: 18775745 DOI: 10.1016/j.dci.2008.07.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 07/25/2008] [Accepted: 07/26/2008] [Indexed: 05/26/2023]
Abstract
Innate immunity is a critical metazoan defense strategy that rapidly detects and neutralizes invading microbes. As the signaling pathways that drive innate immune responses are evolutionarily conserved, there is considerable interest in the characterization of innate immune signaling in genetically tractable models, such as Drosophila melanogaster. Drosophila responds to detection of diamonopimelic-type microbial peptidoglycan through activation of the immune deficiency (Imd) pathway, a signaling pathway with numerous similarities to the mammalian pro-inflammatory TNF pathway. In this manuscript, we focus on a molecular and in vivo characterization of Dnr1, a putative regulator of Imd pathway activity. A previous cell culture RNAi screen indicated that Dnr1 may serve as a negative regulator of the Imd pathway. However, there are no in vivo data to validate this hypothesis and there are scant molecular data to identify the mechanism by which Dnr1 may inhibit the Imd pathway. In this manuscript, we present in vivo data that are consistent with a negative regulatory role for Dnr1 in the Imd pathway. Additionally, we provide molecular data to indicate that Dnr1 inhibits the Imd pathway at the level of the initiator caspase Dredd.
Collapse
Affiliation(s)
- Silvia Guntermann
- Department of Medical Microbiology and Immunology, University of Alberta, Alberta Institute for Viral Immunology, Edmonton AB, T6G 2S2, Canada
| | | | | |
Collapse
|
97
|
Lhocine N, Ribeiro PS, Buchon N, Wepf A, Wilson R, Tenev T, Lemaitre B, Gstaiger M, Meier P, Leulier F. PIMS modulates immune tolerance by negatively regulating Drosophila innate immune signaling. Cell Host Microbe 2008; 4:147-58. [PMID: 18692774 DOI: 10.1016/j.chom.2008.07.004] [Citation(s) in RCA: 175] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2008] [Revised: 06/19/2008] [Accepted: 07/16/2008] [Indexed: 01/20/2023]
Abstract
Metazoans tolerate commensal-gut microbiota by suppressing immune activation while maintaining the ability to launch rapid and balanced immune reactions to pathogenic bacteria. Little is known about the mechanisms underlying the establishment of this threshold. We report that a recently identified Drosophila immune regulator, which we call PGRP-LC-interacting inhibitor of Imd signaling (PIMS), is required to suppress the Imd innate immune signaling pathway in response to commensal bacteria. pims expression is Imd (immune deficiency) dependent, and its basal expression relies on the presence of commensal flora. In the absence of PIMS, resident bacteria trigger constitutive expression of antimicrobial peptide genes (AMPs). Moreover, pims mutants hyperactivate AMPs upon infection with Gram-negative bacteria. PIMS interacts with the peptidoglycan recognition protein (PGRP-LC), causing its depletion from the plasma membrane and shutdown of Imd signaling. Therefore, PIMS is required to establish immune tolerance to commensal bacteria and to maintain a balanced Imd response following exposure to bacterial infections.
Collapse
Affiliation(s)
- Nouara Lhocine
- CNRS, Centre de Génétique Moléculaire, UPR2167, Gif-sur-Yvette, F-91198, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Aggarwal K, Rus F, Vriesema-Magnuson C, Ertürk-Hasdemir D, Paquette N, Silverman N. Rudra interrupts receptor signaling complexes to negatively regulate the IMD pathway. PLoS Pathog 2008; 4:e1000120. [PMID: 18688280 PMCID: PMC2483946 DOI: 10.1371/journal.ppat.1000120] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2008] [Accepted: 07/10/2008] [Indexed: 12/15/2022] Open
Abstract
Insects rely primarily on innate immune responses to fight pathogens. In Drosophila, antimicrobial peptides are key contributors to host defense. Antimicrobial peptide gene expression is regulated by the IMD and Toll pathways. Bacterial peptidoglycans trigger these pathways, through recognition by peptidoglycan recognition proteins (PGRPs). DAP-type peptidoglycan triggers the IMD pathway via PGRP-LC and PGRP-LE, while lysine-type peptidoglycan is an agonist for the Toll pathway through PGRP-SA and PGRP-SD. Recent work has shown that the intensity and duration of the immune responses initiating with these receptors is tightly regulated at multiple levels, by a series of negative regulators. Through two-hybrid screening with PGRP-LC, we identified Rudra, a new regulator of the IMD pathway, and demonstrate that it is a critical feedback inhibitor of peptidoglycan receptor signaling. Following stimulation of the IMD pathway, rudra expression was rapidly induced. In cells, RNAi targeting of rudra caused a marked up-regulation of antimicrobial peptide gene expression. rudra mutant flies also hyper-activated antimicrobial peptide genes and were more resistant to infection with the insect pathogen Erwinia carotovora carotovora. Molecularly, Rudra was found to bind and interfere with both PGRP-LC and PGRP-LE, disrupting their signaling complex. These results show that Rudra is a critical component in a negative feedback loop, whereby immune-induced gene expression rapidly produces a potent inhibitor that binds and inhibits pattern recognition receptors. The innate immune system controls the immediate response to infection. Innate immunity relies on germline encoded receptors, receptors that are present at birth, to recognize germs and trigger a protective response. Invertebrates (i.e., insects) rely on innate immunity to survive in microbial-rich environments, such as rotting fruit. However, uncontrolled innate immune responses are dangerous, leading to severe pathologies like sepsis, inflammatory bowel diseases, and lupus. Therefore, the intensity and duration of the innate immune response is kept in-check by multiple regulatory mechanisms. Here, we have identified a new feedback regulator of the Drosophila (the fruit fly) immune response, which we call Rudra. Using various approaches, we show that in the absence of Rudra the innate immune system is hyper-activated. This elevated immune response leads to better protection against bacterial infection. On the other hand, when present in excess, Rudra prevents the activation of the immune response. Furthermore, we show that Rudra turns off the immune response by binding to the receptors that are responsible for detecting bacteria, thereby preventing downstream responses.
Collapse
Affiliation(s)
- Kamna Aggarwal
- Divison of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Florentina Rus
- Divison of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Christie Vriesema-Magnuson
- Divison of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Deniz Ertürk-Hasdemir
- Divison of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Nicholas Paquette
- Divison of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Neal Silverman
- Divison of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
99
|
Abstract
In the past few years the knowledge of insect defense mechanisms against pathogenic microorganisms and parasites has significantly increased on both the molecular and the organismic level. These investigations have led to new concepts of immune protection also relevant for mammals with the identification of the Toll receptor family as an eminent example. This review provides a brief overview of insect strategies to on the one hand defeat bacterial pathogens while on the other hand cooperating with symbiotic bacteria beneficial for the insects.
Collapse
Affiliation(s)
- Heike Feldhaar
- Lehrstuhl für Soziobiologie und Verhaltensphysiologie, Biozentrum der Universität Würzburg, Am Hubland, D-97074 Würzburg, Germany
| | | |
Collapse
|
100
|
Aggarwal K, Silverman N. Positive and negative regulation of the Drosophila immune response. BMB Rep 2008; 41:267-77. [PMID: 18452646 DOI: 10.5483/bmbrep.2008.41.4.267] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Insects mount a robust innate immune response against a wide array of microbial pathogens. The hallmark of the Drosophila humoral immune response is the rapid production of antimicrobial peptides in the fat body and their release into the circulation. Two recognition and signaling cascades regulate expression of these antimicrobial peptide genes. The Toll pathway is activated by fungal and many Gram-positive bacterial infections, whereas the immune deficiency (IMD) pathway responds to Gram-negative bacteria. Recent work has shown that the intensity and duration of the Drosophila immune response is tightly regulated. As in mammals, hyperactivated immune responses are detrimental, and the proper down-modulation of immunity is critical for protective immunity and health. In order to keep the immune response properly modulated, the Toll and IMD pathways are controlled at multiple levels by a series of negative regulators. In this review, we focus on recent advances identifying and characterizing the negative regulators of these pathways.
Collapse
Affiliation(s)
- Kamna Aggarwal
- Division of Infectious Diseases, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | |
Collapse
|