1
|
Lai Y, Wang S. Epigenetic Regulation in Insect-Microbe Interactions. ANNUAL REVIEW OF ENTOMOLOGY 2025; 70:293-311. [PMID: 39374433 DOI: 10.1146/annurev-ento-022724-010640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Insects have evolved diverse interactions with a variety of microbes, such as pathogenic fungi, bacteria, and viruses. The immune responses of insect hosts, along with the dynamic infection process of microbes in response to the changing host environment and defenses, require rapid and fine-tuned regulation of gene expression programs. Epigenetic mechanisms, including DNA methylation, histone modifications, and noncoding RNA regulation, play important roles in regulating the expression of genes involved in insect immunity and microbial pathogenicity. This review highlights recent discoveries and insights into epigenetic regulatory mechanisms that modulate insect-microbe interactions. A deeper understanding of these regulatory mechanisms underlying insect-microbe interactions holds promise for the development of novel strategies for biological control of insect pests and mitigation of vector-borne diseases.
Collapse
Affiliation(s)
- Yiling Lai
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, China;
| | - Sibao Wang
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, China
- New Cornerstone Science Laboratory, CAS Key Laboratory of Insect Developmental and Evolutionary Biology, CAS Center for Excellence in Molecular Plant Sciences, Shanghai Institute of Plant Physiology and Ecology, Chinese Academy of Sciences (CAS), Shanghai, China;
| |
Collapse
|
2
|
Yao X, Lin L, Ye Z, Huo M, Jin P, Ma F. NF-κB/Relish readjusts miR-100 expression and recovers immune homeostasis in Drosophila melanogaster. INSECT SCIENCE 2024. [PMID: 39688880 DOI: 10.1111/1744-7917.13484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 12/18/2024]
Abstract
The regulation and maintenance of immune homeostasis are essential for animal survival, but the molecular mechanisms are not fully understood. Here, we used the model organism Drosophila melanogaster to uncover a potential mechanism by which the nuclear factor-κB transcription factor Relish and miR-100 cooperatively regulate innate immune homeostasis. We first demonstrated in vitro and in vivo that miR-100 can negatively regulate the immune responses of the Imd pathway by inhibiting the expression of TAK1-associated binding protein 2 (Tab2) gene. Second, we found that Relish, an important transcription factor in the Drosophila Imd pathway, could not only modulate the expressions of antimicrobial peptides (AMPs) to promote immune responses, but also bind to the promoter region of miR-100 and activate its transcription to inhibit immune responses. Third, the dynamic expression of genes profiling indicated that the Relish/miR-100/Tab2 regulatory axis could contribute to innate immune homeostasis in Drosophila. Together, our findings reveal the dual role of Relish in immune regulation, that is, Relish promotes the expression of AMPs to resist pathogen infection in the early immune response, while in the late immune stages, Relish readjusts the expression of miR-100 to negatively control immune responses to avoid excessive immunity thus maintaining immunohomeostasis. Meanwhile, our study provides a new perspective for further understanding the complex regulatory mechanism of immune homeostasis in animals.
Collapse
Affiliation(s)
- Xiaolong Yao
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lu Lin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Zifeng Ye
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Miaomiao Huo
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| |
Collapse
|
3
|
Zhou H, Liu L, Pang Y, Xu Y, Wu J, Ma F, Jin P, Zhou X. Relish-mediated C2H2 zinc finger protein IMZF restores Drosophila immune homeostasis via inhibiting the transcription of Imd/Tak1. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2024; 170:104138. [PMID: 38762126 DOI: 10.1016/j.ibmb.2024.104138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/26/2024] [Accepted: 05/16/2024] [Indexed: 05/20/2024]
Abstract
The dysregulation of intensity and duration in innate immunity can result in detrimental effects on the body, emphasizing the crucial need for precise regulation. However, the intricate and accurate nature of innate immunity implies the existence of numerous undiscovered innate immunomodulators, particularly transcription factors. In this study, we have identified a Drosophila C2H2 zinc finger protein CG18262, named Immune-mediated Zinc Finger protein (IMZF), capable of suppressing immune responses of Imd pathway. Mechanistically, IMZF serves as a transcription factor that represses the expression of Imd and Tak1. Intriguingly, our findings also reveal that Relish, an NF-κB transcription factor, positively regulates the expression of IMZF, consequently inhibiting the activation of Imd and Tak1 to prevent an exaggerated immune response. Additionally, we have elucidated the pivotal role played by the Relish-IMZF-Imd/Tak1 axis in restoring immune homeostasis of Drosophila Imd pathway. In summary, our findings not only unveil a novel C2H2 zinc finger immunoregulatory transcription factor, IMZF, along with its specific mechanism of immune regulation, but also shed light on the dual functionality of Relish in different stages of the immune response by modulating distinct effectors. This discovery provides new insights and enlightenment into the complex regulation of Drosophila innate immunity.
Collapse
Affiliation(s)
- Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Li Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Yujia Pang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Yina Xu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Jing Wu
- Department of Psychology, College of Victoria College, University of Toronto, Toronto, ON, M5R 0A3, Canada
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Xue Zhou
- School of Chemistry and Biological Engineering, Nanjing Normal University Taizhou College, Taizhou, 225300, China.
| |
Collapse
|
4
|
Li Y, Sun Y, Li R, Zhou H, Li S, Jin P. Genetic Screening Revealed the Negative Regulation of miR-310~313 Cluster Members on Imd Pathway during Gram-Negative Bacterial Infection in Drosophila. Genes (Basel) 2024; 15:601. [PMID: 38790230 PMCID: PMC11120675 DOI: 10.3390/genes15050601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/04/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Innate immune response is the first line of host defense against pathogenic microorganisms, and its excessive or insufficient activation is detrimental to the organism. Many individual microRNAs (miRNAs) have emerged as crucial post-transcriptional regulators of immune homeostasis in Drosophila melanogaster. However, the synergistical regulation of miRNAs located within a cluster on the Imd-immune pathway remains obscured. In our study, a genetic screening with 52 transgenic UAS-miRNAs was performed to identify ten miRNAs or miRNA clusters, including the miR310~313 cluster, which may function on Imd-dependent immune responses. The miRNA RT-qPCR analysis showed that the expression of miR-310~313 cluster members exhibited an increase at 6-12 h post E. coli infection. Furthermore, the overexpression of the miR-310~313 cluster impaired the Drosophila survival. And the overexpression of miR-310/311/312 reduced Dpt expression, an indication of Imd pathway induced by Gram-negative bacteria. Conversely, the knockdown of miR-310/311/312 led to increases in Dpt expression. The Luciferase reporter expression assays and RT-qPCR analysis confirmed that miR-310~313 cluster members directly co-targeted and inhibited Imd transcription. These findings reveal that the members of the miR-310~313 cluster synergistically inhibit Imd-dependent immune responses by co-targeting the Imd gene in Drosophila.
Collapse
Affiliation(s)
- Yao Li
- College of Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Yixuan Sun
- College of Plant Protection, Yangzhou University, Yangzhou 225009, China
| | - Ruimin Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China; (R.L.); (H.Z.)
| | - Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China; (R.L.); (H.Z.)
| | - Shengjie Li
- School of Food Science, Nanjing Xiaozhuang University, Nanjing 211171, China;
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China; (R.L.); (H.Z.)
| |
Collapse
|
5
|
Huang Y, Wang T, Jiang C, Li S, Zhou H, Li R. Relish-facilitated lncRNA-CR11538 suppresses Drosophila Imd immune response and maintains immune homeostasis via decoying Relish away from antimicrobial peptide promoters. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2024; 151:105098. [PMID: 37956726 DOI: 10.1016/j.dci.2023.105098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/10/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
Innate immunity plays a crucial role in host defense against pathogen invasion and its strength and duration requires precise control. Long non-coding RNAs (lncRNAs) have become important regulators of innate immunity, yet their roles in Drosophila immune responses remain largely unknown. In this study, we identified that the overexpression of lncRNA-CR11538 inhibits the expression of antimicrobial peptides (AMPs) Dpt and AttA in Drosophila upon Escherichia coli (E. coli) infection, and influences the survival rate of flies after E. cloacae infection. Mechanically, lncRNA-CR11538 decoys Relish away from AMPs promoter region. We further revealed that Relish can promote the transcription of lncRNA-CR11538. After analyzing the dynamic expression profile of lncRNA-CR11538 during Imd immune response, we put forward a hypothesis that in the late stage of Imd immune response, lncRNA-CR11538 can be activated by Relish and further decoy Relish away from the AMPs promoter to suppress excessive immune signal and maintain immune homeostasis. This mechanism we proposed provides insights into the complex regulatory networks controlling immune responses in Drosophila and suggests potential targets for therapeutic intervention in diseases involving dysregulated immune responses.
Collapse
Affiliation(s)
- Yu Huang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, PR China
| | - Tan Wang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, PR China
| | - Chun Jiang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, PR China; Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, Jiangsu, PR China
| | - Shengjie Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, PR China
| | - Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, PR China; Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, 210002, Nanjing, Jiangsu, PR China.
| | - Ruimin Li
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, 455000, PR China.
| |
Collapse
|
6
|
Ren Y, Chen J, Fu S, Bu W, Xue H. Changes in the gut bacterial community affect miRNA profiles in Riptortus pedestris under different rearing conditions. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2023; 48:101135. [PMID: 37688974 DOI: 10.1016/j.cbd.2023.101135] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/04/2023] [Accepted: 08/31/2023] [Indexed: 09/11/2023]
Abstract
Insects possess complex and dynamic gut microbial system, which contributes to host nutrient absorption, reproduction, energy metabolism, and protection against stress. However, there are limited data on interactions of host-gut bacterial microbiota through miRNA (microRNA) regulation in a significant pest, Riptortus pedestris. Here, we performed the 16S rRNA amplicon sequencing and small RNA sequencing from the R. pedestris gut under three environmental conditions and antibiotic treatment, suggesting that we obtained a large amount of reads by assembly, filtration and quality control. The 16S rRNA amplicon sequencing results showed that the abundance and diversity of gut bacterial microbiota were significantly changed between antibiotic treatment and other groups, and they are involved in metabolism and biosynthesis-related function based on functional prediction. Furthermore, we identified different numbers of differentially expressed unigenes (DEGs) and differentially expressed miRNAs (DEMs) based on high-quality mappable reads, which were enriched in various immune-related pathways, including Toll-like receptor, RIG-I-like receptor, NOD-like receptor, JAK/STAT, PI3K/Akt, NF-κB, MAPK signaling pathways, and so forth, using GO and KEGG enrichment analysis. Later on, the identified miRNAs and their target genes in the R. pedestris gut were predicted and randomly selected to construct an interaction network. Finally, our study indicated that alterations in the gut bacterial microbiota are significantly positively or negatively associated with DEMs of the Toll/Imd signaling pathway with Pearson correlation analysis. Taken together, the results of our study lay the foundation for further deeply understanding the interactions between the gut microbiota and immune responses in R. pedestris through miRNA regulation, and provide the new basis for pest management in hemipteran pests.
Collapse
Affiliation(s)
- Yipeng Ren
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| | - Juhong Chen
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| | - Siying Fu
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| | - Wenjun Bu
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| | - Huaijun Xue
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
7
|
Pan W, Yao X, Lin L, Liu X, Jin P, Ma F. The Relish/miR-275/Dredd mediated negative feedback loop is crucial to restoring immune homeostasis of Drosophila Imd pathway. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2023; 162:104013. [PMID: 37804878 DOI: 10.1016/j.ibmb.2023.104013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/13/2023] [Accepted: 09/25/2023] [Indexed: 10/09/2023]
Abstract
The NF-κB/Relish, as a core transcription factor of Drosophila immune deficiency (Imd) pathway, activates the transcriptions of antimicrobial peptides (AMPs) to combat gram-negative bacterial infections, but its role in regulating miRNA expression during immune response has less been reported. We here describe a negative feedback loop of Imd signaling mediated by Relish/miR-275/Dredd that controls Drosophila immune homeostasis after Escherichia coli (E. coli) infection. Our results demonstrate that Relish may directly activate the transcription of miR-275 via binding to its promoter in vitro and vivo, particularly miR-275 further inhibits the expression of Dredd through binding to its 3'UTR to negatively control Drosophila Imd immune response. Remarkably, the ectopic expression of miR-275 significantly reduces Drosophila lifespan. More importantly, our work uncovers a new mechanism by which Relish can flexibly switch its role to maintain Drosophila immune response and homeostasis during infection. Collectively, our study not only reveals the functional duality of Relish in regulating immune response of Drosophila Imd pathway, but also provides a new insight into the maintenance of animal innate immune homeostasis.
Collapse
Affiliation(s)
- Wanwan Pan
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Xiaolong Yao
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Lu Lin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Xiaoqi Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| |
Collapse
|
8
|
Yao X, Ni J, Lin L, Jin P, Ma F. The NF-κB/Relish Activates miR-308 to Negatively Regulate Imd Pathway Immune Signaling in Drosophila. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:591-600. [PMID: 37358278 DOI: 10.4049/jimmunol.2200680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 06/02/2023] [Indexed: 06/27/2023]
Abstract
The strength and duration of the NF-κB signaling response must be tightly modulated to avoid an inadequate or excessive immune response. Relish, a core NF-κB transcription factor of the Drosophila Imd pathway, can control the expression of antimicrobial peptides, including Dpt and AttA, to defend against Gram-negative bacterial infections, but whether Relish may regulate miRNA expression to participate in the immune response remains unclear. In this study, taking advantage of Drosophila S2 cells and different overexpression/knockout/knockdown flies, we first found that Relish could directly activate the expression of miR-308 to negatively regulate the immune response and promote the survival of Drosophila during Enterobacter cloacae infection. Second, our results demonstrated that Relish-mediated expression of miR-308 could suppress target gene Tab2 to attenuate the Drosophila Imd pathway signal during the middle and late stages of the immune response. Third, we detected the dynamic expression patterns of Dpt, AttA, Relish, miR-308, and Tab2 in wild-type flies after E. coli infection, which further revealed that the feedback regulatory loop of Relish-miR-308-Tab2 plays a crucial role in the immune response and homeostasis maintenance of the Drosophila Imd pathway. Overall, our present study not only illustrates an important mechanism by which this Relish-miR-308-Tab2 regulatory axis can negatively control the Drosophila immune response and participate in homeostasis maintenance but also provides new insights into the dynamic regulation of the NF-κB/miRNA expression network of animal innate immunity.
Collapse
Affiliation(s)
- Xiaolong Yao
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Jiajia Ni
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Lu Lin
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics and Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| |
Collapse
|
9
|
Abbas MN, Kausar S, Asma B, Ran W, Li J, Lin Z, Li T, Cui H. MicroRNAs reshape the immunity of insects in response to bacterial infection. Front Immunol 2023; 14:1176966. [PMID: 37153604 PMCID: PMC10161253 DOI: 10.3389/fimmu.2023.1176966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
The interaction between bacteria and insects can significantly impact a wide range of different areas because bacteria and insects are widely distributed around the globe. The bacterial-insect interactions have the potential to directly affect human health since insects are vectors for disease transmission, and their interactions can also have economic consequences. In addition, they have been linked to high mortality rates in economically important insects, resulting in substantial economic losses. MicroRNAs (miRNAs) are types of non-coding RNAs involved in regulating gene expression post-transcriptionally. The length of miRNAs ranges from 19 to 22 nucleotides. MiRNAs, in addition to their ability to exhibit dynamic expression patterns, have a diverse range of targets. This enables them to govern various physiological activities in insects, like innate immune responses. Increasing evidence suggests that miRNAs have a crucial biological role in bacterial infection by influencing immune responses and other mechanisms for resistance. This review focuses on some of the most recent and exciting discoveries made in recent years, including the correlation between the dysregulation of miRNA expression in the context of bacterial infection and the progression of the infection. Furthermore, it describes how they profoundly impact the immune responses of the host by targeting the Toll, IMD, and JNK signaling pathways. It also emphasizes the biological function of miRNAs in regulating immune responses in insects. Finally, it also discusses current knowledge gaps about the function of miRNAs in insect immunity, in addition to areas that require more research in the future.
Collapse
Affiliation(s)
- Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Saima Kausar
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Bibi Asma
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Wenhao Ran
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
| | - Jingui Li
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
| | - Zini Lin
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
| | - Tiejun Li
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Gastrointestinal Vascular Surgery, The Chongqing Ninth People’s Hospital, Chongqing, China
- *Correspondence: Tiejun Li, ; Hongjuan Cui,
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Southwest University, Chongqing, China
- Cancer Center, Medical Research Institute, Southwest University, Chongqing, China
- Jinfeng Laboratory, Chongqing, China
- *Correspondence: Tiejun Li, ; Hongjuan Cui,
| |
Collapse
|
10
|
Lu MY, Chtarbanova S. The role of micro RNAs (miRNAs) in the regulation of Drosophila melanogaster's innate immunity. Fly (Austin) 2022; 16:382-396. [PMID: 36412256 PMCID: PMC9683055 DOI: 10.1080/19336934.2022.2149204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
MicroRNAs (miRNAs) are a class of small non-coding RNAs ~19-22 nt long which post-transcriptionally regulate gene expression. Their ability to exhibit dynamic expression patterns coupled with their wide variety of targets allows miRNAs to regulate many processes, including the innate immune response of Drosophila melanogaster. Recent studies have identified miRNAs in Drosophila which are differentially expressed during infection with different pathogens as well as miRNAs that may affect immune signalling when differentially expressed. This review provides an overview of miRNAswhich have been identified to play a role in the immune response of Drosophila through targeting of the Toll and IMD signalling pathways and other immune processes. It will also explore the role of miRNAs in fine-tuning the immune response in Drosophila and highlight current gaps in knowledge regarding the role of miRNAs in immunity and areas for further research.
Collapse
Affiliation(s)
- Max Yang Lu
- Department of Biological Sciences, the University of Alabama, Tuscaloosa, AL, USA
| | - Stanislava Chtarbanova
- Department of Biological Sciences, the University of Alabama, Tuscaloosa, AL, USA,Center for Convergent Bioscience & Medicine, University of Alabama, Tuscaloosa, AL, USA,Alabama Life Research Institute, University of Alabama, Tuscaloosa, AL, USA,CONTACT Stanislava Chtarbanova Department of Biological Sciences, the University of Alabama, 300, Hackberry Ln, Tuscaloosa, AL-35487, USA
| |
Collapse
|
11
|
Zhou H, Wu S, Liu L, Liu X, Lan S, Jiang J, Yang W, Jin P, Xia X, Ma F. Drosophila Relish-mediated miR-317 expression facilitates immune homeostasis restoration via inhibiting PGRP-LC. Eur J Immunol 2022; 52:1934-1945. [PMID: 36155909 DOI: 10.1002/eji.202250034] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Revised: 09/20/2022] [Accepted: 09/23/2022] [Indexed: 12/13/2022]
Abstract
Innate immunity is the first and essential line for resisting pathogens, and the immune intensity and duration need to be strictly regulated to balance excessive or insufficient immune response. MicroRNAs (miRNAs) are crucial regulators of immune response in Drosophila, yet how immune-related miRNAs are regulated remains poorly understood. Herein, we elucidated that the involvement of miR-317 in NF-κB transcription factor Relish mediated Drosophila Imd pathway in response to Gram-negative (G-) bacteria stimulation. Remarkably, the dynamic expression profiling for immune response indicated that Relish simultaneously enhances the expression of the effector antimicrobial peptide Dpt as well as miR-317 post-infection. Upregulation of miR-317 could further down-regulate the expression of PGRP-LC, thereby forming a feedback in Drosophila Imd pathway to prevent over-activation and restore immune homeostasis. Taken together, our study not only uncovers a novel Relish/miR-317/PGRP-LC regulatory axis to attenuate Drosophila Imd immune response and facilitate immune homeostasis restoration, but also provides vital insights into the complex mechanisms of animal innate immune regulation.
Collapse
Affiliation(s)
- Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China.,Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210002, China
| | - Shanshan Wu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Li Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Xiaoqi Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Siyu Lan
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Jiajun Jiang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Wan Yang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| | - Xinyi Xia
- Institute of Laboratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, 210002, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, P. R. China
| |
Collapse
|
12
|
Liu J, Jin T, Ran L, Zhao Z, Zhu R, Xie G, Bi X. Profiling ATM regulated genes in Drosophila at physiological condition and after ionizing radiation. Hereditas 2022; 159:41. [PMID: 36271387 PMCID: PMC9587650 DOI: 10.1186/s41065-022-00254-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
Background ATM (ataxia-telangiectasia mutated) protein kinase is highly conserved in metazoan, and plays a critical role at DNA damage response, oxidative stress, metabolic stress, immunity, RNA biogenesis etc. Systemic profiling of ATM regulated genes, including protein-coding genes, miRNAs, and long non-coding RNAs, will greatly improve our understanding of ATM functions and its regulation. Results 1) differentially expressed protein-coding genes, miRNAs, and long non-coding RNAs in atm mutated flies were identified at physiological condition and after X-ray irradiation. 2) functions of differentially expressed genes in atm mutated flies, regardless of protein-coding genes or non-coding RNAs, are closely related with metabolic process, immune response, DNA damage response or oxidative stress. 3) these phenomena are persistent after irradiation. 4) there is a cross-talk regulation towards miRNAs by ATM, E2f1, and p53 during development and after irradiation. 5) knock-out flies or knock-down flies of most irradiation-induced miRNAs were sensitive to ionizing radiation. Conclusions We provide a valuable resource of protein-coding genes, miRNAs, and long non-coding RNAs, for understanding ATM functions and regulations. Our work provides the new evidence of inter-dependence among ATM-E2F1-p53 for the regulation of miRNAs. Supplementary Information The online version contains supplementary material available at 10.1186/s41065-022-00254-9.
Collapse
Affiliation(s)
- Jun Liu
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Tianyu Jin
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Lanxi Ran
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Ze Zhao
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Rui Zhu
- College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China
| | - Gangcai Xie
- School of Medicine, Nantong University, Nantong, 226001, China.
| | - Xiaolin Bi
- School of Medicine, Nantong University, Nantong, 226001, China. .,College of Basic Medical Medicine, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
13
|
Zhou H, Wu S, Liu L, Li R, Jin P, Li S. Drosophila Relish Activating lncRNA-CR33942 Transcription Facilitates Antimicrobial Peptide Expression in Imd Innate Immune Response. Front Immunol 2022; 13:905899. [PMID: 35720331 PMCID: PMC9201911 DOI: 10.3389/fimmu.2022.905899] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 05/02/2022] [Indexed: 12/29/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are an emerging class of regulators that play crucial roles in regulating the strength and duration of innate immunity. However, little is known about the regulation of Drosophila innate immunity-related lncRNAs. In this study, we first revealed that overexpression of lncRNA-CR33942 could strengthen the expression of the Imd pathway antimicrobial peptide (AMP) genes Diptericin (Dpt) and Attacin-A (AttA) after infection, and vice versa. Secondly, RNA-seq analysis of lncRNA-CR33942-overexpressing flies post Gram-negative bacteria infection confirmed that lncRNA-CR33942 positively regulated the Drosophila immune deficiency (Imd) pathway. Mechanistically, we found that lncRNA-CR33942 interacts and enhances the binding of NF-κB transcription factor Relish to Dpt and AttA promoters, thereby facilitating Dpt and AttA expression. Relish could also directly promote lncRNA-CR33942 transcription by binding to its promoter. Finally, rescue experiments and dynamic expression profiling post-infection demonstrated the vital role of the Relish/lncRNA-CR33942/AMP regulatory axis in enhancing Imd pathway and maintaining immune homeostasis. Our study elucidates novel mechanistic insights into the role of lncRNA-CR33942 in activating Drosophila Imd pathway and the complex regulatory interaction during the innate immune response of animals.
Collapse
Affiliation(s)
- Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Shanshan Wu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Li Liu
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ruimin Li
- College of Biology and Food Engineering, Anyang Institute of Technology, Anyang, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Shengjie Li
- Jiangsu Provincial Key Construction Laboratory of Special Biomass Byproduct Resource Utilization, School of Food Science, Nanjing Xiaozhuang University, Nanjing, China
| |
Collapse
|
14
|
Ko HJ, Jo YH, Patnaik BB, Park KB, Kim CE, Keshavarz M, Jang HA, Lee YS, Han YS. IKKγ/NEMO Is Required to Confer Antimicrobial Innate Immune Responses in the Yellow Mealworm, Tenebrio Molitor. Int J Mol Sci 2020; 21:ijms21186734. [PMID: 32937897 PMCID: PMC7555931 DOI: 10.3390/ijms21186734] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/31/2020] [Accepted: 09/07/2020] [Indexed: 12/14/2022] Open
Abstract
IKKγ/NEMO is the regulatory subunit of the IκB kinase (IKK) complex, which regulates the NF-κB signaling pathway. Within the IKK complex, IKKγ/NEMO is the non-catalytic subunit, whereas IKKα and IKKβ are the structurally related catalytic subunits. In this study, TmIKKγ was screened from the Tenebrio molitor RNA-Seq database and functionally characterized using RNAi screening for its role in regulating T. molitor antimicrobial peptide (AMP) genes after microbial challenges. The TmIKKγ transcript is 1521 bp that putatively encodes a polypeptide of 506 amino acid residues. TmIKKγ contains a NF-κB essential modulator (NEMO) and a leucine zipper domain of coiled coil region 2 (LZCC2). A phylogenetic analysis confirmed its homology to the red flour beetle, Tribolium castaneum IKKγ (TcIKKγ). The expression of TmIKKγ mRNA showed that it might function in diverse tissues of the insect, with a higher expression in the hemocytes and the fat body of the late-instar larvae. TmIKKγ mRNA expression was induced by Escherichia coli, Staphylococcus aureus, and Candida albicans challenges in the whole larvae and in tissues such as the hemocytes, gut and fat body. The knockdown of TmIKKγ mRNA significantly reduced the survival of the larvae after microbial challenges. Furthermore, we investigated the tissue-specific induction patterns of fourteen T. molitor AMP genes in TmIKKγ mRNA-silenced individuals after microbial challenges. In general, the mRNA expression of TmTenecin1, -2, and -4; TmDefensin1 and -2; TmColeoptericin1 and 2; and TmAttacin1a, 1b, and 2 were found to be downregulated in the hemocytes, gut, and fat body tissues in the TmIKKγ-silenced individuals after microbial challenges. Under similar conditions, TmRelish (NF-κB transcription factor) mRNA was also found to be downregulated. Thus, TmIKKγ is an important factor in the antimicrobial innate immune response of T. molitor.
Collapse
Affiliation(s)
- Hye Jin Ko
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.J.K.); (Y.H.J.); (K.B.P.); (C.E.K.); (M.K.); (H.A.J.)
| | - Yong Hun Jo
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.J.K.); (Y.H.J.); (K.B.P.); (C.E.K.); (M.K.); (H.A.J.)
| | - Bharat Bhusan Patnaik
- School of Biotech Sciences, Trident Academy of Creative Technology (TACT), Chandrasekharpur, Bhubaneswar, Odisha 751024, India;
- P.G. Department of Bio-Sciences and Bio-Technology, Fakir Mohan University, Nuapadhi, Balasore, Odisha 756089, India
| | - Ki Beom Park
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.J.K.); (Y.H.J.); (K.B.P.); (C.E.K.); (M.K.); (H.A.J.)
| | - Chang Eun Kim
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.J.K.); (Y.H.J.); (K.B.P.); (C.E.K.); (M.K.); (H.A.J.)
| | - Maryam Keshavarz
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.J.K.); (Y.H.J.); (K.B.P.); (C.E.K.); (M.K.); (H.A.J.)
| | - Ho Am Jang
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.J.K.); (Y.H.J.); (K.B.P.); (C.E.K.); (M.K.); (H.A.J.)
| | - Yong Seok Lee
- School of Biotechnology and Life Sciences, College of Natural Sciences, Soonchunhyang University, 22 Soonchunhyangro, Shinchang-Myeon, Asan, Chungchungnam-do 31538, Korea;
| | - Yeon Soo Han
- Department of Applied Biology, Institute of Environmentally-Friendly Agriculture (IEFA), College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Korea; (H.J.K.); (Y.H.J.); (K.B.P.); (C.E.K.); (M.K.); (H.A.J.)
- Correspondence: ; Tel.: +82-62-530-2072
| |
Collapse
|
15
|
Li R, Zhou H, Jia C, Jin P, Ma F. Drosophila Myc restores immune homeostasis of Imd pathway via activating miR-277 to inhibit imd/Tab2. PLoS Genet 2020; 16:e1008989. [PMID: 32810129 PMCID: PMC7455005 DOI: 10.1371/journal.pgen.1008989] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/28/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Drosophila Myc (dMyc), as a broad-spectrum transcription factor, can regulate the expression of a large number of genes to control diverse cellular processes, such as cell cycle progression, cell growth, proliferation and apoptosis. However, it remains largely unknown about whether dMyc can be involved in Drosophila innate immune response. Here, we have identified dMyc to be a negative regulator of Drosophila Imd pathway via the loss- and gain-of-function screening. We demonstrate that dMyc inhibits Drosophila Imd immune response via directly activating miR-277 transcription, which further inhibit the expression of imd and Tab2-Ra/b. Importantly, dMyc can improve the survival of flies upon infection, suggesting inhibiting Drosophila Imd pathway by dMyc is vital to restore immune homeostasis that is essential for survival. Taken together, our study not only reports a new dMyc-miR-277-imd/Tab2 axis involved in the negative regulation of Drosophila Imd pathway, and provides a new insight into the complex regulatory mechanism of Drosophila innate immune homeostasis maintenance.
Collapse
Affiliation(s)
- Ruimin Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Chaolong Jia
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
- * E-mail: (PJ); (FM)
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, China
- * E-mail: (PJ); (FM)
| |
Collapse
|
16
|
González-González A, Wayne ML. Immunopathology and immune homeostasis during viral infection in insects. Adv Virus Res 2020; 107:285-314. [PMID: 32711732 DOI: 10.1016/bs.aivir.2020.06.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Organisms clear infections by mounting an immune response that is normally turned off once the pathogens have been cleared. However, sometimes this immune response is not properly or timely arrested, resulting in the host damaging itself. This immune dysregulation may be referred to as immunopathology. While our knowledge of immune and metabolic pathways in insects, particularly in response to viral infections, is growing, little is known about the mechanisms that regulate this immune response and hence little is known about immunopathology in this important and diverse group of organisms. In this chapter we focus both on documenting the molecular mechanisms described involved in restoring immune homeostasis in insects after viral infections and on identifying potential mechanisms for future investigation. We argue that learning about the immunopathological consequences of an improperly regulated immune response in insects will benefit both insect and human health.
Collapse
Affiliation(s)
| | - Marta L Wayne
- Department of Biology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
17
|
Ma L, Liu L, Zhao Y, Yang L, Chen C, Li Z, Lu Z. JNK pathway plays a key role in the immune system of the pea aphid and is regulated by microRNA-184. PLoS Pathog 2020; 16:e1008627. [PMID: 32584915 PMCID: PMC7343183 DOI: 10.1371/journal.ppat.1008627] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/08/2020] [Accepted: 05/13/2020] [Indexed: 12/21/2022] Open
Abstract
Different from holometabolous insects, the hemipteran species such as pea aphid Acyrthosiphon pisum exhibit reduced immune responses with the absence of the genes coding for antimicrobial peptide (AMP), immune deficiency (IMD), peptidoglycan recognition proteins (PGRPs), and other immune-related molecules. Prior studies have proved that phenoloxidase (PO)-mediated melanization, hemocyte-mediated phagocytosis, and reactive oxygen species (ROS) participate in pea aphid defense against bacterial infection. Also, the conserved signaling, Jun N-terminal kinase (JNK) pathway, has been suggested to be involved in pea aphid immune defense. However, the precise role of the JNK signaling, its interplay with other immune responses and its regulation in pea aphid are largely unknown. In this study, using in vitro biochemical assays and in vivo bioassays, we demonstrated that the JNK pathway regulated hemolymph PO activity, hydrogen peroxide concentration and hemocyte phagocytosis in bacteria infected pea aphids, suggesting that the JNK pathway plays a central role in regulating immune responses in pea aphid. We further revealed the JNK pathway is regulated by microRNA-184 in response to bacterial infection. It is possible that in common the JNK pathway plays a key role in immune system of hemipteran insects and microRNA-184 regulates the JNK pathway in animals.
Collapse
Affiliation(s)
- Li Ma
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Lu Liu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Yujie Zhao
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Lei Yang
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Caihua Chen
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhaofei Li
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling, Shaanxi, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail:
| |
Collapse
|
18
|
Li R, Huang Y, Zhang Q, Zhou H, Jin P, Ma F. The miR-317 functions as a negative regulator of Toll immune response and influences Drosophila survival. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 95:19-27. [PMID: 30708026 DOI: 10.1016/j.dci.2019.01.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 01/26/2019] [Accepted: 01/27/2019] [Indexed: 06/09/2023]
Abstract
The miR-317 has been revealed to involve in the reproductive response and the larval ovary morphogenesis of Drosophila. However, whether the miR-317 can also regulate Drosophila innate immune responses, which remains unclear to date. Here we have verified that miR-317 can directly target the 3'UTR of Dif-Rc to down-regulate the expression levels of AMP Drs to negatively control Drosophila Toll immune response in vivo and vitro. Specially, the Dif is an important transcription factor of Toll pathway with four transcripts (Dif-Ra, Dif-Rb, Dif-Rc and Dif-Rd). Our results show that miR-317 only targets to Dif-Rc, but not Dif-Ra/b/d, implying that miRNAs can regulate different isoforms of an alternative splicing gene to fine tune immune responses and maintain homeostasis in post-transcriptional level. Furthermore, we have demonstrated that the miR-317 sponge can restore the expression levels of Drs and Dif-Rc at mRNA and protein levels. Remarkably, during Gram-positive bacterial infection, the overexpressed miR-317 flies have poor survival outcome, whereas the knockout miR-317 flies have favorable survival compared to the control group, respectively, suggesting that the miR-317 might play a key role in Drosophila survival. Taken together, our current works not only reveal an innate immune function and a novel regulation pattern of miR-317, but also provide a new insight into the underlying molecular mechanisms of immunity disorder influencing on Drosophila survival.
Collapse
Affiliation(s)
- Ruimin Li
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Yu Huang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Qi Zhang
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Hongjian Zhou
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China
| | - Ping Jin
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics & Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing, 210046, China.
| |
Collapse
|
19
|
Portet A, Pinaud S, Chaparro C, Galinier R, Dheilly NM, Portela J, Charriere GM, Allienne JF, Duval D, Gourbal B. Sympatric versus allopatric evolutionary contexts shape differential immune response in Biomphalaria / Schistosoma interaction. PLoS Pathog 2019; 15:e1007647. [PMID: 30893368 PMCID: PMC6443186 DOI: 10.1371/journal.ppat.1007647] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 04/01/2019] [Accepted: 02/19/2019] [Indexed: 12/22/2022] Open
Abstract
Selective pressures between hosts and their parasites can result in reciprocal evolution or adaptation of specific life history traits. Local adaptation of resident hosts and parasites should lead to increase parasite infectivity/virulence (higher compatibility) when infecting hosts from the same location (in sympatry) than from a foreign location (in allopatry). Analysis of geographic variations in compatibility phenotypes is the most common proxy used to infer local adaptation. However, in some cases, allopatric host-parasite systems demonstrate similar or greater compatibility than in sympatry. In such cases, the potential for local adaptation remains unclear. Here, we study the interaction between Schistosoma and its vector snail Biomphalaria in which such discrepancy in local versus foreign compatibility phenotype has been reported. Herein, we aim at bridging this gap of knowledge by comparing life history traits (immune cellular response, host mortality, and parasite growth) and molecular responses in highly compatible sympatric and allopatric Schistosoma/Biomphalaria interactions originating from different geographic localities (Brazil, Venezuela and Burundi). We found that despite displaying similar prevalence phenotypes, sympatric schistosomes triggered a rapid immune suppression (dual-RNAseq analyses) in the snails within 24h post infection, whereas infection by allopatric schistosomes (regardless of the species) was associated with immune cell proliferation and triggered a non-specific generalized immune response after 96h. We observed that, sympatric schistosomes grow more rapidly. Finally, we identify miRNAs differentially expressed by Schistosoma mansoni that target host immune genes and could be responsible for hijacking the host immune response during the sympatric interaction. We show that despite having similar prevalence phenotypes, sympatric and allopatric snail-Schistosoma interactions displayed strong differences in their immunobiological molecular dialogue. Understanding the mechanisms allowing parasites to adapt rapidly and efficiently to new hosts is critical to control disease emergence and risks of Schistosomiasis outbreaks. Schistosomiasis, the second most widespread human parasitic disease after malaria, is caused by helminth parasites of the genus Schistosoma. More than 200 million people in 74 countries suffer from the pathological, and societal consequences of this disease. To complete its life cycle, the parasite requires an intermediate host, a freshwater snail of the genus Biomphalaria for its transmission. Given the limited options for treating Schistosoma mansoni infections in humans, much research has focused on developing methods to control transmission by its intermediate snail host. Biomphalaria glabrata. Comparative studies have shown that infection of the snail triggers complex cellular and humoral immune responses resulting in significant variations in parasite infectivity and snail susceptibility, known as the so-called polymorphism of compatibility. However, studies have mostly focused on characterizing the immunobiological mechanisms in sympatric interactions. Herein we used a combination of molecular and phenotypic approaches to compare the effect of infection in various sympatric and allopatric evolutionary contexts, allowing us to better understand the mechanisms of host-parasite local adaptation. Learning more about the immunobiological interactions between B. glabrata and S. mansoni could have important socioeconomic and public health impacts by changing the way we attempt to eradicate parasitic diseases and prevent or control schistosomiasis in the field.
Collapse
Affiliation(s)
- Anaïs Portet
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
| | - Silvain Pinaud
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
| | - Cristian Chaparro
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
| | - Richard Galinier
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
| | - Nolwenn M. Dheilly
- School of Marine and Atmospheric Sciences, Stony Brook University, Stony Brook, New York, United States of America
| | - Julien Portela
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
| | - Guillaume M. Charriere
- Interactions Hôtes-Pathogènes-Environnements (IHPE), UMR 5244, CNRS, Ifremer, Université de Perpignan Via Domitia, Université de Montpellier, Montpellier, France
| | - Jean-François Allienne
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
| | - David Duval
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
| | - Benjamin Gourbal
- Univ. Perpignan Via Domitia, Interactions Hôtes Pathogènes Environnements UMR 5244, CNRS, IFREMER, Univ. Montpellier, Perpignan, France
- * E-mail:
| |
Collapse
|
20
|
Wei G, Sun L, Li R, Li L, Xu J, Ma F. Dynamic miRNA-mRNA regulations are essential for maintaining Drosophila immune homeostasis during Micrococcus luteus infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 81:210-224. [PMID: 29198775 DOI: 10.1016/j.dci.2017.11.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/15/2017] [Accepted: 11/29/2017] [Indexed: 06/07/2023]
Abstract
Pathogen bacteria infections can lead to dynamic changes of microRNA (miRNA) and mRNA expression profiles, which may control synergistically the outcome of immune responses. To reveal the role of dynamic miRNA-mRNA regulation in Drosophila innate immune responses, we have detailedly analyzed the paired miRNA and mRNA expression profiles at three time points during Drosophila adult males with Micrococcus luteus (M. luteus) infection using RNA- and small RNA-seq data. Our results demonstrate that differentially expressed miRNAs and mRNAs represent extensively dynamic changes over three time points during Drosophila with M. luteus infection. The pathway enrichment analysis indicates that differentially expressed genes are involved in diverse signaling pathways, including Toll and Imd as well as orther signaling pathways at three time points during Drosophila with M. luteus infection. Remarkably, the dynamic change of miRNA expression is delayed by compared to mRNA expression change over three time points, implying that the "time" parameter should be considered when the function of miRNA/mRNA is further studied. In particular, the dynamic miRNA-mRNA regulatory networks have shown that miRNAs may synergistically regulate gene expressions of different signaling pathways to promote or inhibit innate immune responses and maintain homeostasis in Drosophila, and some new regulators involved in Drosophila innate immune response have been identified. Our findings strongly suggest that miRNA regulation is a key mechanism involved in fine-tuning cooperatively gene expressions of diverse signaling pathways to maintain innate immune response and homeostasis in Drosophila. Taken together, the present study reveals a novel role of dynamic miRNA-mRNA regulation in immune response to bacteria infection, and provides a new insight into the underlying molecular regulatory mechanism of Drosophila innate immune responses.
Collapse
Affiliation(s)
- Guanyun Wei
- Laboratory for Comparative Genomics and Bioinformatics, Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Lianjie Sun
- Laboratory for Comparative Genomics and Bioinformatics, Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Ruimin Li
- Laboratory for Comparative Genomics and Bioinformatics, Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Lei Li
- Laboratory for Comparative Genomics and Bioinformatics, Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China; Laboratory of Intelligent Computation, School of Computer Science, Nanjing Normal University, Nanjing 210046, China
| | - Jiao Xu
- Laboratory for Comparative Genomics and Bioinformatics, Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China
| | - Fei Ma
- Laboratory for Comparative Genomics and Bioinformatics, Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Science, Nanjing Normal University, Nanjing 210046, China.
| |
Collapse
|