Next Article in Journal
A Fatal Case of Metastatic Pulmonary Calcification during the Puerperium
Previous Article in Journal
Shedding Light on the Role of Na,K-ATPase as a Phosphatase during Matrix-Vesicle-Mediated Mineralization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Intestinal Immune Defense System in Insects

Department of Entomology, College of Plant Protection, South China Agricultural University, Guangzhou 510642, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(23), 15132; https://doi.org/10.3390/ijms232315132
Submission received: 1 November 2022 / Revised: 29 November 2022 / Accepted: 29 November 2022 / Published: 1 December 2022
(This article belongs to the Section Molecular Immunology)

Abstract

:
Over a long period of evolution, insects have developed unique intestinal defenses against invasion by foreign microorganisms, including physical defenses and immune responses. The physical defenses of the insect gut consist mainly of the peritrophic matrix (PM) and mucus layer, which are the first barriers to pathogens. Gut microbes also prevent the colonization of pathogens. Importantly, the immune-deficiency (Imd) pathways produce antimicrobial peptides to eliminate pathogens; mechanisms related to reactive oxygen species are another important pathway for insect intestinal immunity. The janus kinase/STAT signaling pathway is involved in intestinal immunity by producing bactericidal substances and regulating tissue repair. Melanization can produce many bactericidal active substances into the intestine; meanwhile, there are multiple responses in the intestine to fight against viral and parasitic infections. Furthermore, intestinal stem cells (ISCs) are also indispensable in intestinal immunity. Only the coordinated combination of the intestinal immune defense system and intestinal tissue renewal can effectively defend against pathogenic microorganisms.

1. Introduction

Feeding in insects is an important source of intestinal microorganisms, with large numbers of food-derived microorganisms entering the gut of insects every day, which need to be removed to ensure a healthy gut. Due to the long-term interactions between microorganisms and the gut, the latter has developed a series of defense systems (Figure 1 and Table 1).
The physical structure of the insect gut is the first line of defense against invasion by foreign pathogens [1]. The peritrophic membrane (PM) is one of the important members of the insect intestinal physical defense system, which is mainly composed of chitin and protein [1,37]. The mucus composed of glycosylated proteins is another key physical structure [38]. In addition, the acidic areas of the gut and the intestinal epithelial cells act as natural barriers against pathogens [1,37,38,39]. At the same time, the differentiation in intestinal stem cells provides a continuous impetus for the renewal of the natural barrier [40,41]. The combination of the intestinal physical structure and stem cell differentiation results in a complex and effective physical defense system.
Numerous studies have shown that the immune defense response (Figure 1) is rapidly initiated when intestinal epithelial cells are attacked by pathogens [9,42,43,44,45,46,47], and an imbalance in gut homeostasis can also induce an immune response [48]. The insect gut activates the immune-deficiency (Imd) pathway to produce antimicrobial peptides (AMPs) [49,50,51], which mainly destroy Gram-negative microorganisms. Mechanisms related to reactive oxygen species (ROS) provide a very important line of defense in the midgut immune response and can kill ROS-sensitive microorganisms [52,53]. The janus kinase (JAK)/STAT signaling pathway is also involved in intestinal immune regulation through the production of antimicrobial peptides and the regulation of intestinal stem cell (ISC) differentiation [54] The melanization regulated by prophenoloxidase (proPO) produces a variety of active antimicrobial substances that also have an important role in insect intestinal immunity [55]. Therefore, the intestinal immune defense response is of particular importance in the resistance and elimination of foreign pathogens and in maintaining the stability of the internal environment of the intestinal microbial microbiome.
In this paper, we reviewed the progress of research on insect gut defenses against exogenous pathogens in terms of physical defenses, gut microbes, the gut immune system, and intestinal stem cell differentiation.

2. Structure and Function of the Insect Intestine

The insect gut is a tubular structure that connects mouth to anus and runs throughout the entire body cavity [56]. The gut of most adult insects is composed of three structurally, functionally, and developmentally distinct primary domains: the foregut, the midgut, and the hindgut [39,57].
The foregut is located at the most anterior part and includes the pharynx, esophagus, crop, and cardia flap, which have the functions of ingestion, swallowing, chewing, and temporary storage of food [58]. Meanwhile the crop of most insects also has digestive, detoxification and immunological functions, such as in Diptera [45,59]. The cardia flap is the dividing point between the foregut and midgut [55] and can effectively prevent bacteria and particles larger than 0.2 μm from entering the midgut and hindgut by mechanically breaking down food, thereby facilitating host gut defense against insect pathogens [60]. It has also been shown that the cardia in Drosophila and Glossina morsitans encodes and expresses AMP genes [61,62].
The midgut consists of the peritrophic membrane, the intestinal wall cell layer, the basal membrane, the circular and the longitudinal muscle in order from the inside out [63,64,65,66]. There are four types of intestinal wall cells: enterocytes (ECs) secreting digestive enzymes and absorbing nutrients, enteroendocrine cells (EECs) secreting hormones, intestinal stem cells (ISCs) with differentiation functions, and enteroblasts (EBs) with limited differentiation [66,67,68]. The primary function of the midgut is similar to that of the mammalian stomach, which secretes a large number of digestive enzymes and is an important place for digesting food and absorbing nutrients, as well as being the center of the immune response in the insect gut [64,69,70,71].
The hindgut is connected to Malpighian tubules at the anterior end and consists of the pyloric valve, ileum, colon, and rectum. Its main function is to eliminate the digested food waste from the intestine and recycle water and inorganic salts from the residue [72,73].

3. The Physical Defense System of the Insect Intestine

3.1. Peritrophic Membrane

The foregut and hindgut of insects are protected by a tightly arranged semi-permeable membrane structure of chitin and protein, known as the peritrophic membrane (PM) [56]. However, the midgut of Hemiptera does not have PM and instead contains a microvillous membrane, the perimicrovillar membrane (PMM) [74]. It is the first physical barrier of the insect intestinal immune system, which prevents damage caused by pathogens, food particles and bacterial toxins ingested by insects and coming into direct contact with intestinal epithelial cells [75,76]. Therefore, the PM is a defense outpost for pathogenic microbial infections that occur in invertebrates via food. As the PM has a role in isolating pathogens, its thickness and integrity are particularly important in defense. Drosocrystallin (dcy) protein is a major component of the PM, and mortality is significantly increased in dcy mutant Drosophila following intestinal infection with Pseudomonas entomophila [38,77,78]. Moreover, the integrity of the PM may be regulated through the Wnt (Wingless/Integrated) signaling pathway in tsetse flies [79]. It has also been shown that the integrity of the PM is a key factor in the regulation of intestinal homeostasis during changiing in the gut microbiota loads in Anopheles coluzzii [80]. In addition, transglutaminase (TG) in Drosophila crosslinks with fructose crystal glycoproteins situated on the PM to form stable fibrous structures that more effectively defend against infection by P. entomophila in the intestine [77].

3.2. Mucus

In many insects, there is a physical barrier between the intestinal epithelium and the intestinal contents, similar to the mucus layer of the vertebrate digestive tract, auxiliary to the PM [2]. Prior to the discovery of the insect gut mucus layer, it was thought that the PM was a structure in the insect gut analogous to the vertebrate mucus layer in protecting cells from acid damage. The mucus is made of mucus-forming proteins (Mf-mucins), which are highly glycosylated proteins found on the surface of epithelial cells lining the respiratory, digestive, and urogenital tracts of vertebrates [39,81]. The physical barrier protects intestinal epithelial cells from infection, dehydration, and physical and chemical damage and facilitates the passage of food through the intestine [81].
More than 30 genes encoding mucin analogs have been identified in Drosophila, but little is known about the function of these genes and the role of the encoded mucins [81]. Transcriptome analysis Drosophila and Bactrocera dorsalis with oral bacterial infection showed that the expression of genes regulating the PM and mucus layer composition was significantly upregulated, suggesting that the insect gut builds two physical barriers in response to pathogenic bacterial invasion [9,82].

3.3. Other Physical and Chemical Barriers

The outermost layer of the enterocytes of most insects is also covered with a layer of tightly arranged microvilli, mainly composed of actin [83]. When Serratia marcescens and Bacillus thuringiensis invade the intestines of B. dorsalis and Leptinotarsa decemlineata, respectively, the microvilli are abnormally shed as the infection progresses [9,84]. Meanwhile, the genes Big Bang and myosin IB (Myo1B) encode septal junction strength and microvilli structures [44,85].
In addition to the physical barrier of the PM and mucus, there are also physicochemical factors in the intestinal lumen that can degrade pathogens and prevent them from passing through the intestinal epithelium, such as the acidic region of the intestinal lumen [86], digestive enzymes [87], lysozymes [88,89,90], and peptidoglycan hydrolase [91].

4. Gut Microbes

The insect gut is an important interface between the host and the external environment, inhabiting a large number of microorganisms. Insect gut microbes play an important role in host nutrition, detoxification, growth, and activation of immune responses [92,93,94]. Meanwhile, in natural open environments, insect feeding is the source of complex and diverse foreign microorganisms including pathogenic microorganisms. Therefore, maintaining the homeostasis of the intestinal microbiome, resisting and eliminating foreign pathogens is crucial for the physiological ecology of insect growth, development, and reproduction.
The gut commensal microbiota of insects is involved in immune defense response, either directly or indirectly. It has been shown that the Drosophila intestinal clearance response to viruses relies on gut commensal bacteria to initiate the Imd signaling pathway as a means of activating the ERK signaling pathway in the gut [95]. Recent studies have also shown that the long red cone midge Rhodnius prolixus significantly enhances the immune response of the gut upon antibiotic treatment to remove intestinal bacteria by recolonizing S. marcescens or Rhodococcus rhodnii [96,97]. Ordinarily, intestinal commensal bacteria can protect the host by altering physiological functions, pH, and digestive enzyme levels in the gut and can also eliminate pathogens by competing with them for space and nutrients or by producing antibacterial substances [59]. In mosquitoes, ROS, metabolites, small peptides, and proteins secreted by gut bacteria may directly influence the transmission and development of pathogens [98]. A recent study also confirmed that the Bombyx mori intestinal commensal Enterococcus faecalis LX10 significantly reduced the germination rate and effectiveness of infection by the microsporidium Nosema bombycis [99]. Meanwhile, intestinal commensal bacteria activate the Imd–Relish immune pathway to balance the regeneration of intestinal epithelial cells caused by pathogenic bacteria [100]. The aposymbiotic insects succumbed significantly faster than conventionally reared insects upon bacterial infection in Rhynchophorus ferrugineus [101] The endosymbiotic mosquito Wolbachia significantly reduces the infection rate of dengue virus and Plasmodium falciparum, thus blocking the transmission of dengue fever and malaria [102].
Conversely, some intestinal bacteria secrete substances that inhibit the intestinal immune response and increase the insecticidal activity of pathogenic bacteria. It has been reported that the gut microbe Citrobacter freundii of L. decemlineata accelerates sepsis caused by B. thuringiensis [91]. Moreover, the pathogenic fungus Beauveria bassiana interacts with intestinal microbiota to accelerate the death of mosquitoes [103].

5. Intestinal Immunity Response

When pathogenic bacteria break through the first line (physical defense system) of the insect gut, the host produces AMPs/ROS-active substances with antimicrobial activity for clearance. Meanwhile, when the gut is attacked by viruses or parasites, it likewise elicits a strong host rejection response, such as activation of the JAK/STAT signaling pathway or the melanization response. In this section, we describe the modulation of multiple pathways activated by pathogens.

5.1. Imd Signaling Pathway

AMPs are produced in cellular immunity by two main signaling pathways: Imd and Toll, but it has been shown that AMPs in the insect gut are mainly derived from the Imd signaling pathway [104]. The peptidoglycan recognition protein family (PGRPs), which is subdivided into the transmembrane protein receptor PGRP-LC and the cytoplasmic receptor PGRP-LE, recognizes diaminoylpeptidoglycan (DAP-PGN) or peptidoglycan monomers (TCT) of pathogenic bacteria to activate the Imd signaling pathway in the insect gut (Figure 2) [105,106,107]. PGRP-SD is located upstream of PGRP-LC, and binding to DAP-PGN enhances the recognition signal of PGRP-LC [108]. Recognition of peptidoglycan by PGRPs transmits signals downstream to activate the expression of TAK1 and IKK, and the N-terminus of the NF-κB-like protein Relish is transferred to the nucleus to initiate the expression of AMP genes [3,43,52].
Excess AMPs are detrimental to both the intestine and its normal flora, so the Imd signaling pathway in the gut is regulated by multiple precise and complex negative regulatory mechanisms (Figure 2) [50]. These negative regulators fall into two main categories, one targeting activators of the Imd signaling pathway and one targeting key genes of the Imd signaling pathway. PGRPs with amidase activity, including PGRP-LB, PGRP-SB1, PGRP-SB2, PGRP-SC1a, PGRP-SC1b, PGRP-SC, and PGRPC-SC2, cleave peptidoglycan in the intestine to reduce the source of stimulation of the Imd signaling pathway [10,109,110]. Moreover, Rudra, PIRK, and PGRP-LF and PGRP-LF target the Imd signaling pathway receptors and decrease the number of peptidoglycan recognition receptors [111,112,113]. There are also negative regulatory protein targets that inhibit the Imd signaling pathway, including Dnr1 (Defense repressor 1), which inhibits DREDD activity [114,115]; Caspar, which inhibits the Imd signaling pathway by cleaving relish dependent on DREDD-generated Relish and thus is negatively regulated [116,117,118]; Trabid regulates TAK1 levels [119]; SkpA, which is a subunit of SCF-E3 ubiquitin ligase, targets Relish [120,121]; transcriptional repressors including Caudal [122,123], Oct1 homolog Bbin [124], PP4 (Protein Phosphatase 4) [125] and Myc [126].

5.2. Duox–ROS Defense System

In addition to AMPs, ROS also have antibacterial activity in the insect gut [5,127]. Activation of the intestinal nicotinamide adenine dinucleotide phosphate oxidase Duox by pathogenic microorganisms produces ROS that can directly destroy pathogenic bacteria, fungi, or plasmodia [127], thus the DUOX-ROS system plays an important role in insect intestinal immunity. In addition to its involvement in the clearance of pathogenic microorganisms, the Duox-ROS system plays an essential role in maintaining intestinal homeostasis in B. dorsalis [128]. Meanwhile, a recent study also showed that serotonin in the gut of B. dorsalis and Aedes aegypti affects the homeostasis of gut microbiota by regulating the expression of Duox [129].
Many studies have shown that insect intestinal Duox activation produces ROS in two directions, one regulating Duox gene expression in the nucleus and the other activating Duox enzyme activity (Figure 3A). Meanwhile, Duox gene expression is also regulated by two distinct pathways: the MEKK1–MKK3–p38–ATF2 signaling pathway regulates Duox expression in the nucleus [59], and the cell membrane protein Mesh induces changes in Duox expression through an Arrestin-mediated phosphorylation cascade reaction of MAPK JNK/ERK [130]. Furthermore, the MEKK1–MKK3–p38–ATF2 signaling pathway is also dependent on the activation of the PGRPs downstream of peptidoglycan in the Imd signaling pathway, which does not affect the enzymatic activity of Duox [131]. The specific ligand that activates Duox enzymatic activity is uracil, which is secreted by most pathogenic bacteria but not intestinal commensal microbes [132]. It regulates the Hedgehog (Hh) signaling pathway while being recognized by the G-protein coupled receptor (GPCR), activating the formation of Cad99C/PLCβ/PKC endosomes, leading to Ca2+ release from the endoplasmic reticulum and activating the enzymatic activity of Duox [133,134].
Peptidoglycan-dependent activation of the Duox transcriptional pathway is negatively regulated by p38 activation, which itself is regulated by PLCβ, Calcineurin B (CanB) and MAP kinase phosphatase 3 (MKP3) (Figure 3A) [135]. This negative regulation ensures that transcriptional Duox is activated only when stimulated by large amounts of peptidoglycan, thus being able to protect the normal proliferation of the intestinal flora. ROS scavenging mechanisms exist in the insect gut, as excess ROS cause oxidative stress damage to intestinal epithelial cells. ROS enzymes are mainly involved in regulating normal levels of ROS that maintain normal oxidative reactions in the intestine and avoid cell damage by the excess of ROS such as catalase [127], long-oxide dehydrogenase, thioredoxin peroxidase, and glutathione peroxidase [12]. The Nrf2/Keap1 signaling pathway activates the production of the abovementioned ROS enzymes when intestinal epithelial cells are exposed to oxidative stress [136,137]. Upon an invasion of exogenous pathogens in the gut, Duox regulates ROS in order to clear the overexpressed ROS in the immune response by various peroxidases in vivo to maintain their levels within the threshold of host damage.

5.3. JAK/STAT Signaling Pathway

In conjugation with the Duox and Imd signaling pathways which are two major complementary immune defense pathways in the insect gut, there are other pathways that activate AMPs to participate in the immune defense response, such as the activation of the midgut JAK/STAT pathway that induces Drosomycin expression to defend against fungal invasion [138,139,140,141]. The pathway is conserved throughout biological evolution and plays an important role in insect innate immunity [141,142]. Numerous studies have revealed its involvement in intestinal immune responses, such as resistance to viral infections [143,144,145,146] and resistance to fungal and bacterial infections [147,148].
Regulation of the JAK/STAT signaling pathway is mediated by a variety of cytokines that regulate many important biological processes such as immune regulation, cell proliferation, differentiation, and apoptosis [149]. The JAK/STAT signaling pathway is activated by the binding of secreted ligands to receptors, leading to the aggregation of JAK and the activation of STAT by phosphorylation and subsequent translocation to the nucleus to regulate the expression of target genes. In contrast to the multiple JAK/STAT combination pathways in mammals, only one typical pathway has been identified in Drosophila [150,151]. It has three main components: the receptor Domeless [152,153]; the JAK Hopscotch [154] and the transcription factor STAT92E [155,156]; Domeless is regulated by an unpaired (Upd) family of three secreted proteins, Upd1, Upd2, and Upd3 [157]. The pathway is also regulated by a number of negative feedback regulators, including Socs36E, Ptp61F, and Wdp [158,159,160].
Numerous studies have revealed an important role of the JAK/STAT pathway in the insect intestinal immune response (Figure 3). Starting with the observation of phosphorylation and translocation of STAT92E in Anopheles gambiae infected with enteric bacteria [161], there has been growing evidence of its involvement in the insect intestinal immune response. Intestinal infection with the pathogens results in damage to the intestinal epithelium, leading to the release of the ligands Upd2 and Upd3 that activate the expression of Drosomycin (Figure 3B) [138,162]. Contact between common microbes and intestinal commensal microorganisms also activates the JAK/STAT pathway to enhance immunity in the gut [163]. Furthermore, it has also been shown that the production of Upd3 and activation of the JAK/STAT pathway in the gut must also depend on the oxidative burst of Duox (Figure 3A) [164]. Meanwhile, the JAK/STAT pathway has a crucial role in host defense against viral infection according to activating the expression of IFN-stimulated genes (ISGs), which are a group of secreted proteins that play key roles in innate immunity [165,166].

5.4. Melanization in the Insect Gut

Insect intestinal immunity also includes a melanization response activated by the propheoloxidase activating system (proPO system) (Figure 1) [64,167]. It is triggered by the activation of proPO enzymes through the protein hydrolysis of a series of clip domain serine proteases, which produce polyphenol oxidase (PPO) that oxidize phenolic compounds such as tyrosine and dopa to quinone, which is further converted to melanin [168,169]. The activation process of melanization produces many antimicrobial active substances with bactericidal properties that enable the clearance of pathogenic bacteria in intestinal infections [170].
The melanization response contributes to wound healing by isolating dying pathogens from surrounding tissues and is therefore primarily associated with death or tissue damage [40,171,172]. Current research on the origin of PPO has led to the following findings: the midgut PPO of adult mosquitoes probably originates from the hemolymph [171,172,173]; the presence of PPO is not detected in the midgut of the silkworm, but is found in the hindgut, which is therefore the center of melanization [174]. Pathogenic microorganisms enter the hindgut from the midgut and before being eliminated from the body, there is a PPO bactericidal response in the intestine, which becomes the last line of immune defense of the insect gut.
However, B. thuringiensis can inhibit the melanization reaction by producing Bt protein crystal protein 1Ab (Cry1Ab), leading to an increase in intestinal flora density in the hindgut of silkworms [175]. Microbes in the midgut of Helicoverpa armigera also mediate the downregulation of the antiviral factor PPO to promote baculovirus infection [176].

5.5. Intestinal Immunity against Parasitic and Viral Infections

Inclusive of bacterial infections in the insect gut, there are also microsporidia and viruses that parasitize the intestinal epithelium and cause infections [177]. As with pathogenic bacteria, the entry of a virus or parasite into the insect’s gut exposes it to the physical defenses of the gut, the first line of defense, as well as activating the immune response of the intestinal epithelium.
Infection of B. mori larvae with the silkworm microsporidian N. bombycis activates the midgut Toll pathway and the JAK-STAT pathway to produce antimicrobial peptides, and melanization in the hemolymph of silkworm larvae was also found to be inhibited [178]. This suggests that there is information transfer between the hemolymph and the gut when the insect gut is infected. When the intestinal parasite melanizes in mosquitoes, it is retained throughout the life cycle of the insect [172]. Moreover, after the intestinal tract of the bumblebee Bombus inptiens is infected with the parasite Crithidia bombi, the central nervous system pathway will be destroyed, and cognition and behavior change, indicating that there is brain–gut axis communication [179].
Viruses enter intestinal epithelial cells through virus receptors in intestinal microvilli, causing a host immune response [180]. Bmlipase-1 (Bm serine protease-1) [181], BmSP-2 (Bm serine protease-2) [70], and alkaline trypsin in the gut of the silkworm have strong antiviral activity against the nucleopolyhedrovirus (MNPV) [182]. Analysis of the midgut transcriptome of silkworms infected with Bombyx mori cytoplasmic polyhedrosis virus (BmCPV) revealed increased expression of the serine protease inhibitor Serpin-5 [183]. Additionally, infection of Autographa californica with AcMNPV regulated expression of p35 and inhibited apoptosis, suggesting that host cell apoptosis is also a strategy to combat virus infection [184]. Meanwhile, studies of insect intestinal resistance to viral infestation have shown that the Imd signaling pathway is also involved in the antiviral response [96]. When viruses enter the Drosophila intestine, the PGRP-LC receptor on the intestinal epithelial cell membrane recognizes peptidoglycans of the microorganis and activates the Imd signaling pathway, which initiates NF-κB signaling and activates the expression of Pvf2 ligands, thereby activating ERK antiviral activity in the intestine through the binding of Pvf2 ligands to the PVR receptor [95].

6. Gut Renewal

The intestinal epithelium of adult insect, such as vertebrates, is a highly regenerative tissue that rapidly self-renews in response to changing inputs from nutrition, gut microbiota, ingested toxins, and signals from other organs [185]. The regeneration of insect intestinal epithelial cells is controlled by the proliferation and differentiation of ISCs, which play a replenishing role in intestinal defense against pathogen invasion (Figure 3B).
It has been reported that intestinal epithelial cells of adult Drosophila are controlled by the proliferation and differentiation of ISCs, thereby controlling the rate of regenerative renewal [65]. When pathogenic bacteria or toxic substances invade the insect intestine and cause acute damage to intestinal cells, Drosophila regulates intestinal epithelial cell structure and function by altering the rate of ISC proliferation and differentiation to repair intestinal damage [186]. Signaling pathways such as JAK/STAT, EGFR, Hippo, and JNK have been reported to be involved in regulating ISC differentiation [162,187,188,189].
Interestingly, recent studies suggested that secondary metabolites of enteric microbiota also activate the differentiation of ISCs [190]. Meanwhile, gut microbes promote ISC differentiation into enterocytes (ECs) via the microbial pattern recognition pathway Imd/Relish, whereas pathogenic bacteria promote ISC differentiation into enteroendocrine cells (EE) via the JAK/STAT signaling pathway [100]. It has been found that an increase in the number of gut bacteria in Imd-mutant Drosophila caused an accelerated rate of ISC division, whereas ISC proliferation and differentiation was inhibited in Duox-mutant Drosophila, suggesting that the ROS molecules produced by Duox are key factors in the induction of ISC division [82]. Excess ROS in the intestinal immune response damages intestinal epithelial cells, which induces ISC proliferation and differentiation (Figure 3A).
Therefore, intestinal epithelial cell differentiation is very importantly linked to the intestinal immune system, and only the immune response together with intestinal stem cell differentiation can work together to fight pathogenic bacteria quickly and efficiently [191].

7. Conclusions

Exogenous pathogens that enter the insect gut are intercepted and eliminated by a variety of factors. Upon pathogen invasion, intestinal epithelial cells are protected by different means such as the PM, mucus layer, acidic zone, and various digestive enzymes. These cells can recognize pathogenic bacteria and activate the production of bactericidal effector molecules such as AMPs and ROS in the intestine, thus eliminating harmful microorganisms. Nevertheless, Imd and JAK/STAT signaling pathways produce the AMPs, Duox/Mesh, which mediate the expression of ROS and melanization cascades involving numerous diverse bactericidal substances that also play important roles in intestinal immunity. Enhancing the ability of intestinal epithelial cells to proliferate and differentiate is also a way to defend against foreign pathogens. The entire intestinal immune defense system helps the intestine to fight off pathogens very effectively.
An immune homeostatic mechanism exists in the insect gut that ensures effective defense against foreign pathogen invasion while protecting the gut from damage caused by immune overload. This homeostasis, developed during the long evolution of insects, has been a hot topic of research in recent years. Many studies have demonstrated multiple mechanisms in host insects to ensure a moderate immune response, ensuring an appropriate immune response to pathogenic bacteria and the regulation of gut flora homeostasis while avoiding an overexcited immune response.
The study of insect intestinal immune responses has provided a theoretical basis for opening up new technologies for agricultural pest control. In recent years, many studies have focused on the killing power of bacterial functional molecules, while less research has been carried out on the disordered killing power of insect intestinal immune homeostatic mechanisms. The lethal role of disordered intestinal immune homeostasis in pest control has broad application prospects, and the use of genetic engineering to modify insects while exploring new homeostatic mechanisms will be one of the research hotspots in coming years.
Although much has been achieved in the study of the insect intestinal immune system, many open questions remain. The Imd signaling pathway and Duox-ROS defense system have been shown to activate ISC differentiation and enhance the renewal rate of intestinal epithelial cells in addition to removing pathogens. This demonstrates that the regenerative renewal capacity of intestinal epithelial cells is particularly important for insects to defend themselves against pathogenic microorganisms. At the same time, there may be additional signaling pathways that jointly regulate ISC differentiation. It further remains unknown or what measures insects use to coordinate the direction of ISC differentiation and competition for differentiation. We believe that this will be the focus of future research. In addition, research on the brain–gut axis (the regulatory system for two-way communication between the nervous and gastrointestinal systems) has received attention in recent years. Communications between the intestinal immune system and the nervous system in insects lead to changes in activity and behavior, which may also provide more possibilities for the future application of intestinal probiotics in human medicine.

Author Contributions

Y.Q. and Y.X. conceived the project. T.Z. wrote the manuscript and drew the pictures. S.J. revised all the manuscript. Y.Q. and Y.X. contributed to revise the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by National Key R&D Program of China (Grant No. 2021YFD1000500) and the National Key Research and Development Program of China (2021YFC2600404).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Acknowledgments

We sincerely thank lab members for their valuable advice in preparing this review.

Conflicts of Interest

The authors declared there is no conflict between the authors.

References

  1. Hegedus, D.; Erlandson, M.; Gillott, C.; Toprak, U. New insights into peritrophic matrix synthesis, architecture, and function. Annu. Rev. Entomol. 2009, 54, 285–302. [Google Scholar] [CrossRef] [PubMed]
  2. Dias, R.O.; Cardoso, C.; Pimentel, A.C.; Damasceno, T.F.; Ferreira, C.; Terra, W.R. The roles of mucus-forming mucins, peritrophins and peritrophins with mucin domains in the insect midgut. Insect Mol. Biol. 2018, 27, 46–60. [Google Scholar] [CrossRef] [PubMed]
  3. Kleino, A.; Silverman, N. Regulation of the Drosophila Imd pathway by signaling amyloids. Insect Biochem. Mol. 2019, 108, 16–23. [Google Scholar] [CrossRef] [PubMed]
  4. Bahuguna, S.; Atilano, M.; Glittenberg, M.; Lee, D.; Arora, S.; Wang, L.; Zhou, J.; Redhai, S.; Boutros, M.; Ligoxygakis, P. Bacterial recognition by PGRP-SA and downstream signalling by Toll/DIF sustain commensal gut bacteria in Drosophila. PLoS Genet. 2022, 18, e1009992. [Google Scholar]
  5. Ha, E.M.; Oh, C.T.; Bae, Y.S.; Lee, W.J. A direct role for dual oxidase in Drosophila gut immunity. Science 2005, 310, 847–850. [Google Scholar] [CrossRef]
  6. Lourido, F.; Quenti, D.; Salgado-Canales, D.; Tobar, N. Domeless receptor loss in fat body tissue reverts insulin resistance induced by a high-sugar diet in Drosophila melanogaster. Sci. Rep. 2021, 11, 3263. [Google Scholar] [CrossRef] [PubMed]
  7. Iatsenko, I.; Boquete, J.P.; Lemaitre, B. Microbiota-derived lactate activates production of Reactive Oxygen Species by the Intestinal NADPH Oxidase Nox and shortens Drosophila lifespan. Immunity 2018, 49, 929–942.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Troha, K.; Buchon, N. Methods for the study of innate immunity in Drosophila melanogaster. Wiley Interdiscip. Rev. Dev. Biol. 2019, 8, e344. [Google Scholar] [CrossRef]
  9. Zeng, T.; Bai, X.; Liu, Y.L.; Li, J.F.; Lu, Y.Y.; Qi, Y.X. Intestinal responses of the oriental fruit fly Bactrocera dorsalis (Hendel) after ingestion of an entomopathogenic bacterium strain. Pest Manag. Sci. 2020, 76, 653–664. [Google Scholar] [CrossRef]
  10. Zhang, P.; Yao, Z.; Bai, S.; Zhang, H. The negative regulative roles of BdPGRPs in the Imd signaling pathway of Bactrocera dorsalis. Cells 2022, 11, 152. [Google Scholar] [CrossRef]
  11. Liu, S.H.; Wei, D.; Yuan, G.R.; Jiang, H.B.; Dou, W.; Wang, J.J. Antimicrobial peptide gene cecropin-2 and defensin respond to peptidoglycan infection in the female adult of oriental fruit fly, Bactrocera dorsalis (Hendel). Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2017, 206, 1–7. [Google Scholar] [CrossRef] [PubMed]
  12. Molina-Cruz, A.; Dejong, R.J.; Charles, B.; Gupta, L.; Kumar, S.; Jaramillo-Gutierrez, G.; Barillas-Mury, C. Reactive oxygen species modulate Anopheles gambiae immunity against bacteria and plasmodium. J. Biol. Chem. 2008, 283, 3217–3223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Magalhaes, T. What is the association of heme aggregates with the peritrophic matrix of adult female mosquitoes? Parasites Vectors 2014, 7, 362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Dias, R.O.; Cardoso, C.; Leal, C.S.; Ribeiro, A.F.; Ferreira, C.; Terra, W.R. Domain structure and expression along the midgut and carcass of peritrophins and cuticle proteins analogous to peritrophins in insects with and without peritrophic membrane. J. Insect. Physiol. 2019, 114, 1–9. [Google Scholar] [CrossRef]
  15. Meister, S.; Agianian, B.; Turlure, F.; Relogio, A.; Morlais, I.; Kafatos, F.C.; Christophides, G.K. Anopheles gambiae PGRPLC-mediated defense against bacteria modulates infections with malaria parasites. PLoS Pathog. 2009, 5, e1000542. [Google Scholar] [CrossRef] [Green Version]
  16. Kumar, S.; Molina-Cruz, A.; Gupta, L.; Rodrigues, J.; Barillas-Mury, C. Peroxidase/dual oxidase system modulates midgut epithelial immunity in Anopheles gambiae. Science 2010, 327, 1644–1648. [Google Scholar] [CrossRef] [Green Version]
  17. Horton, A.A.; Wang, B.; Camp, L.; Price, M.S.; Arshi, A.; Nagy, M.; Nadler, S.A.; Faeder, J.R.; Luckhart, S. The mitogen-activated protein kinome from Anopheles gambiae: Identification, phylogeny and functional characterization of the ERK, JNK and p38 MAP kinases. BMC Genom. 2011, 12, 574. [Google Scholar] [CrossRef] [Green Version]
  18. Zhong, X.W.; Wang, X.H.; Tan, X.; Xia, Q.Y.; Xiang, Z.H.; Zhao, P. Identification and molecular characterization of a chitin deacetylase from Bombyx mori peritrophic membrane. Int. J. Mol. Sci. 2014, 15, 1946–1961. [Google Scholar] [CrossRef] [Green Version]
  19. Nesa, J.; Sadat, A.; Buccini, D.F.; Kati, A.; Mandal, A.K.; Franco, O.L. Antimicrobial peptides from Bombyx mori: A splendid immune defense response in silkworms. RSC Adv. 2020, 10, 512–523. [Google Scholar] [CrossRef] [Green Version]
  20. Lin, J.H.; Yu, X.Q.; Wang, Q.; Tao, X.P.; Li, J.Y.; Zhang, S.S.; Xia, X.F.; You, M.S. Immune responses to Bacillus thuringiensis in the midgut of the diamondback moth, Plutella xylostella. Dev. Comp. Immunol. 2020, 107, 103661. [Google Scholar] [CrossRef]
  21. Sarauer, B.L.; Gillott, C.; Hegedus, D. Characterization of an intestinal mucin from the peritrophic matrix of the diamondback moth, Plutella xylostella. Insect Mol. Biol. 2003, 12, 333–343. [Google Scholar] [CrossRef] [PubMed]
  22. Lin, J.H.; Xia, X.F.; Yu, X.Q.; Shen, J.H.; Li, Y.; Lin, H.L.; Tang, S.S.; Vasseur, L.; You, M.S. Gene expression profiling provides insights into the immune mechanism of Plutella xylostella midgut to microbial infection. Gene 2018, 647, 21–30. [Google Scholar] [CrossRef] [PubMed]
  23. Silva, C.P.; Silva, J.R.; Vasconcelos, F.F.; Petretski, M.D.A.; DaMatta, R.A.; Ribeiro, A.F.; Terra, W.R. Occurrence of midgut perimicrovillar membranes in paraneopteran insect orders with comments on their function and evolutionary significance. Arthropod. Struct. Dev. 2004, 33, 139–148. [Google Scholar] [CrossRef]
  24. Gerardo, N.M.; Altincicek, B.; Anselme, C.; Atamian, H.; Barribeau, S.M.; de Vos, M.; Duncan, E.J.; Evans, J.D.; Gabaldon, T.; Ghanim, M.; et al. Immunity and other defenses in pea aphids, Acyrthosiphon pisum. Genome Biol. 2010, 11, R21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zhou, X.; Peng, L.Y.; Wang, Z.C.; Wang, W.; Zhu, Z.; Huang, X.H.; Chen, L.B.; Song, Q.S.; Bao, Y.Y. Identification of novel antimicrobial peptides from rice planthopper, Nilaparvata lugens. Insect Biochem. Mol. Biol. 2019, 113, 103215. [Google Scholar] [CrossRef] [PubMed]
  26. Bao, Y.Y.; Qu, L.Y.; Zhao, D.; Chen, L.B.; Jin, H.Y.; Xu, L.M.; Cheng, J.A.; Zhang, C.X. The genome- and transcriptome-wide analysis of innate immunity in the brown planthopper, Nilaparvata lugens. BMC Genom. 2013, 14, 160. [Google Scholar] [CrossRef] [Green Version]
  27. Liu, X.; Li, J.; Guo, W.; Li, R.; Zhao, D.; Li, X. A new type I peritrophic membrane protein from larval Holotrichia oblita (Coleoptera: Melolonthidae) binds to chitin. Int. J. Mol. Sci. 2014, 15, 6831–6842. [Google Scholar] [CrossRef] [Green Version]
  28. Xiao, R.; Wang, X.; Xie, E.; Ji, T.; Li, X.; Muhammad, A.; Yin, X.; Hou, Y.; Shi, Z. An IMD-like pathway mediates the intestinal immunity to modulate the homeostasis of gut microbiota in Rhynchophorus ferrugineus Olivier (Coleoptera: Dryophthoridae). Dev. Comp. Immunol. 2019, 97, 20–27. [Google Scholar] [CrossRef]
  29. Muhammad, A.; Habineza, P.; Wang, X.; Xiao, R.; Ji, T.; Hou, Y.; Shi, Z. Spatzle Homolog-mediated Toll-like pathway regulates innate immune responses to maintain the homeostasis of gut microbiota in the Red Palm Weevil, Rhynchophorus ferrugineus Olivier (Coleoptera: Dryophthoridae). Front. Microbiol. 2020, 11, 846. [Google Scholar] [CrossRef]
  30. Yin, A.; Pan, L.; Zhang, X.; Wang, L.; Yin, Y.; Jia, S.; Liu, W.; Xin, C.; Liu, K.; Yu, X.; et al. Transcriptomic study of the red palm weevil Rhynchophorus ferrugineus embryogenesis. Insect Sci. 2015, 22, 65–82. [Google Scholar] [CrossRef]
  31. Szymas, B.; Langowska, A.; Kazimierczak-Baryczko, M. Histological structure of the midgut of honey bees (Apis mellifera L.) fed pollen substitutes fortified with probiotics. J. Apic. Sci. 2012, 56, 5–12. [Google Scholar] [CrossRef]
  32. Maruscakova, I.C.; Schusterova, P.; Bielik, B.; Toporcak, J.; Bilikova, K.; Mudronova, D. Effect of application of probiotic pollen suspension on immune response and gut microbiota of honey bees (Apis mellifera). Probiotics Antimicrob. Proteins 2020, 12, 929–936. [Google Scholar] [CrossRef] [PubMed]
  33. Azzouz-Olden, F.; Hunt, A.; De Grandi-Hoffman, G. Transcriptional response of honey bee (Apis mellifera) to differential nutritional status and Nosema infection. BMC Genom. 2018, 19, 628. [Google Scholar] [CrossRef] [PubMed]
  34. Rutter, L.; Carrillo-Tripp, J.; Bonning, B.C.; Cook, D.; Toth, A.L.; Dolezal, A.G. Transcriptomic responses to diet quality and viral infection in Apis mellifera. BMC Genom. 2019, 20, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Emery, O.; Schmidt, K.; Engel, P. Immune system stimulation by the gut symbiont Frischella perrara in the honey bee (Apis mellifera). Mol. Ecol. 2017, 26, 2576–2590. [Google Scholar] [CrossRef]
  36. Whitehorn, P.R.; Tinsley, M.C.; Brown, M.J.F.; Darvill, B.; Goulson, D. Genetic diversity, parasite prevalence and immunity in wild bumblebees. Proc. R. Soc. B Biol. Sci. 2011, 278, 1195–1202. [Google Scholar] [CrossRef]
  37. Kuraishi, T.; Binggeli, O.; Opota, O.; Buchon, N.; Lemaitre, B. Genetic evidence for a protective role of the peritrophic matrix against intestinal bacterial infection in Drosophila melanogaster. Proc. Natl. Acad. Sci. USA 2011, 108, 15966–15971. [Google Scholar] [CrossRef] [Green Version]
  38. Miguel-Aliaga, I.; Jasper, H.; Lemaitre, B. Anatomy and physiology of the digestive tract of Drosophila melanogaster. Genetics 2018, 210, 357–396. [Google Scholar] [CrossRef] [Green Version]
  39. Ma, E.H.; Zhu, Y.B.; Liu, Z.W.; Wei, T.Y.; Wang, P.H.; Cheng, G. Interaction of viruses with the insect intestine. Annu. Rev. Virol. 2021, 8, 115–131. [Google Scholar] [CrossRef]
  40. Jin, Y.; Patel, P.H.; Kohlmaier, A.; Pavlovic, B.; Zhang, C.; Edgar, B.A. Intestinal stem cell pool regulation in Drosophila. Stem Cell Rep. 2017, 8, 1479–1487. [Google Scholar] [CrossRef] [Green Version]
  41. Zwick, R.K.; Ohlstein, B.; Klein, O.D. Intestinal renewal across the animal kingdom: Comparing stem cell activity in mouse and Drosophila. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 316, G313–G322. [Google Scholar] [CrossRef]
  42. Paredes, J.C.; Welchman, D.P.; Poidevin, M.; Lemaitre, B. Negative regulation by amidase PGRPs shapes the Drosophila antibacterial response and protects the fly from innocuous infection. Immunity 2011, 35, 770–779. [Google Scholar] [CrossRef] [Green Version]
  43. Bosco-Drayon, V.; Poidevin, M.; Boneca, I.G.; Narbonne-Reveau, K.; Royet, J.; Charroux, B. Peptidoglycan sensing by the receptor PGRP-LE in the Drosophila gut induces immune responses to infectious bacteria and tolerance to microbiota. Cell Host Microbe 2012, 12, 153–165. [Google Scholar] [CrossRef] [Green Version]
  44. Bonnay, F.; Cohen-Berros, E.; Hoffmann, M.; Kim, S.Y.; Boulianne, G.L.; Hoffmann, J.A.; Matt, N.; Reichhart, J.M. Big bang gene modulates gut immune tolerance in Drosophila. Proc. Natl. Acad. Sci. USA 2013, 110, 2957–2962. [Google Scholar] [CrossRef] [Green Version]
  45. Stoffolano, J.G.; Haselton, A.T. The Adult Dipteran crop: A unique and overlooked organ. Annu. Rev. Entomol. 2013, 58, 205–225. [Google Scholar] [CrossRef]
  46. Chen, K.K.; Lu, Z.Q. Immune responses to bacterial and fungal infections in the silkworm, Bombyx mori. Dev. Comp. Immunol. 2018, 83, 3–11. [Google Scholar] [CrossRef]
  47. Wu, S.; Zhang, X.F.; Chen, X.M.; Cao, P.S.; Beerntsen, B.T.; Ling, E.J. BmToll9, an Arthropod conservative Toll, is likely involved in the local gut immune response in the silkworm, Bombyx mori. Dev. Comp. Immunol. 2010, 34, 93–96. [Google Scholar] [CrossRef] [PubMed]
  48. Buchon, N.; Broderick, N.A.; Lemaitre, B. Gut homeostasis in a microbial world: Insights from Drosophila melanogaster. Nat. Rev. Microbiol. 2013, 11, 615–626. [Google Scholar] [CrossRef] [Green Version]
  49. Simoes, M.L.; Goncalves, L.; Silveira, H. Hemozoin activates the innate immune system and reduces Plasmodium berghei infection in Anopheles gambiae. Parasites Vectors 2015, 8, 12. [Google Scholar] [CrossRef] [Green Version]
  50. Liu, W.; Wang, Y.; Zhou, J.; Zhang, Y.; Ma, Y.; Wang, D.; Jiang, Y.; Shi, S.; Qin, L. Peptidoglycan recognition proteins regulate immune response of Antheraea pernyi in different ways. J. Invertebr. Pathol. 2019, 166, 107204. [Google Scholar] [CrossRef]
  51. Chang, M.M.; Wang, Y.H.; Yang, Q.T.; Wang, X.L.; Wang, M.; Raikhel, A.S.; Zou, Z. Regulation of antimicrobial peptides by juvenile hormone and its receptor, Methoprene-tolerant, in the mosquito Aedes aegypti. Insect Biochem. Mol. 2021, 128, 103509. [Google Scholar] [CrossRef] [PubMed]
  52. Leulier, F.; Parquet, C.; Pili-Floury, S.; Ryu, J.H.; Caroff, M.; Lee, W.J.; Mengin-Lecreulx, D.; Lemaitre, B. The Drosophila immune system detects bacteria through specific peptidoglycan recognition. Nat. Immunol. 2003, 4, 478–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Jang, S.; Mergaert, P.; Ohbayashi, T.; Ishigami, K.; Shigenobu, S.; Itoh, H.; Kikuchi, Y. Dual oxidase enables insect gut symbiosis by mediating respiratory network formation. Proc. Natl. Acad. Sci. USA 2021, 118, e2020922118. [Google Scholar] [CrossRef] [PubMed]
  54. Hoffmann, J.A. The immune response of Drosophila. Nature 2003, 426, 33–38. [Google Scholar] [CrossRef]
  55. Lu, A.R.; Zhang, Q.L.; Zhang, J.; Yang, B.; Wu, K.; Xie, W.; Luan, Y.X.; Ling, E.J. Insect prophenoloxidase: The view beyond immunity. Front. Physiol. 2014, 5, 252. [Google Scholar] [CrossRef] [Green Version]
  56. Buchon, N.; Osman, D. All for one and one for all: Regionalization of the Drosophila intestine. Insect Biochem. Mol. 2015, 67, 2–8. [Google Scholar] [CrossRef]
  57. Buchon, N.; Osman, D.; David, F.P.A.; Fang, H.Y.; Boquete, J.P.; Deplancke, B.; Lemaitre, B. Morphological and molecular characterization of adult midgut compartmentalization in Drosophila. Cell Rep. 2013, 3, 1725–1738. [Google Scholar] [CrossRef] [Green Version]
  58. Stoffolano, J.G. Fly foregut and transmission of microbes. Adv. Insect. Physiol. 2019, 57, 27–95. [Google Scholar]
  59. Bai, S.; Yao, Z.; Raza, M.F.; Cai, Z.; Zhang, H. Regulatory mechanisms of microbial homeostasis in insect gut. Insect Sci. 2021, 28, 286–301. [Google Scholar] [CrossRef]
  60. Lanan, M.C.; Rodrigues, P.A.P.; Agellon, A.; Jansma, P.; Wheeler, D.E. A bacterial filter protects and structures the gut microbiome of an insect. ISME J. 2016, 10, 1866–1876. [Google Scholar] [CrossRef] [Green Version]
  61. Tzou, P.; Ohresser, S.; Ferrandon, D.; Capovilla, M.; Reichhart, J.M.; Lemaitre, B.; Hoffmann, J.A.; Imler, J.L. Tissue-specific inducible expression of antimicrobial peptide genes in Drosophila surface epithelia. Immunity 2000, 13, 737–748. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Hu, C.; Aksoy, S. Innate immune responses regulate trypanosome parasite infection of the tsetse fly Glossina morsitans. Mol. Microbiol. 2006, 60, 1194–1204. [Google Scholar] [CrossRef] [PubMed]
  63. Broderick, N.A.; Buchon, N.; Lemaitre, B. Microbiota-induced changes in Drosophila melanogaster host gene expression and gut morphology. mBio 2014, 5, e01117-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Wu, K.; Yang, B.; Huang, W.; Dobens, L.; Song, H.; Ling, E. Gut immunity in Lepidopteran insects. Dev. Comp. Immunol. 2016, 64, 65–74. [Google Scholar] [CrossRef]
  65. Micchelli, C.A.; Perrimon, N. Evidence that stem cells reside in the adult Drosophila midgut epithelium. Nature 2006, 439, 475–479. [Google Scholar] [CrossRef]
  66. Ohlstein, B.; Spradling, A. The adult Drosophila posterior midgut is maintained by pluripotent stem cells. Nature 2006, 439, 470–474. [Google Scholar] [CrossRef]
  67. Ohlstein, B.; Kai, T.; Decotto, E.; Spradling, A. The stem cell niche: Theme and variations. Curr. Opin. Cell. Biol. 2004, 16, 693–699. [Google Scholar] [CrossRef]
  68. Ohlstein, B.; Spradling, A. Multipotent Drosophila intestinal stem cells specify daughter cell fates by differential notch signaling. Science 2007, 315, 988–992. [Google Scholar] [CrossRef] [Green Version]
  69. Yao, Z.; Cai, Z.; Ma, Q.; Bai, S.; Wang, Y.; Zhang, P.; Guo, Q.; Gu, J.; Lemaitre, B.; Zhang, H. Compartmentalized PGRP expression along the dipteran Bactrocera dorsalis gut forms a zone of protection for symbiotic bacteria. Cell Rep. 2022, 41, 111523. [Google Scholar] [CrossRef]
  70. Nakazawa, H.; Tsuneishi, E.; Ponnuvel, K.M.; Furukawa, S.; Asaoka, A.; Tanaka, H.; Ishibashi, J.; Yamakawa, M. Antiviral activity of a serine protease from the digestive juice of Bombyx mori larvae against nucleopolyhedrovirus. Virology 2004, 321, 154–162. [Google Scholar] [CrossRef] [Green Version]
  71. Janeh, M.; Osman, D.; Kambris, Z. Comparative analysis of midgut regeneration capacity and resistance to oral infection in three disease-vector mosquitoes. Sci. Rep. 2019, 9, 14556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Murakami, R.; Shiotsuki, Y. Ultrastructure of the hindgut of Drosophila larvae, with special reference to the domains identified by specific gene expression patterns. J. Morphol. 2001, 248, 144–150. [Google Scholar] [CrossRef] [PubMed]
  73. Lengyel, J.A.; Iwaki, D.D. It takes guts: The Drosophila hindgut as a model system for organogenesis. Dev. Biol. 2002, 243, 1–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Peng, X.; Zha, W.; He, R.; Lu, T.; Zhu, L.; Han, B.; He, G. Pyrosequencing the midgut transcriptome of the brown planthopper, Nilaparvata lugens. Insect Mol. Biol. 2011, 20, 745–762. [Google Scholar] [CrossRef] [PubMed]
  75. Kuraishi, T.; Hori, A.; Kurata, S. Host-microbe interactions in the gut of Drosophila melanogaster. Front. Physiol. 2013, 4, 375. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Villegas-Ospina, S.; Merritt, D.J.; Johnson, K.N. Physical and chemical barriers in the larval midgut confer developmental resistance to virus infection in Drosophila. Viruses 2021, 13, 894. [Google Scholar] [CrossRef]
  77. Shibata, T.; Maki, K.; Hadano, J.; Fujikawa, T.; Kitazaki, K.; Koshiba, T.; Kawabata, S. Crosslinking of a peritrophic matrix protein protects gut epithelia from bacterial exotoxins. PLoS Pathog. 2015, 11, e1005244. [Google Scholar] [CrossRef] [Green Version]
  78. Prakash, A.; Monteith, K.M.; Vale, P.F. Mechanisms of damage prevention, signalling and repair impact disease tolerance. Proc. Biol. Sci. 2022, 289, 20220837. [Google Scholar] [CrossRef]
  79. Aksoy, S. Tsetse peritrophic matrix influences for trypanosome transmission. J. Insect Physiol. 2019, 118, 103919. [Google Scholar] [CrossRef]
  80. Rodgers, F.H.; Gendrin, M.; Wyer, C.A.S.; Christophides, G.K. Microbiota-induced peritrophic matrix regulates midgut homeostasis and prevents systemic infection of malaria vector mosquitoes. PLoS Pathog. 2017, 13, e1006391. [Google Scholar] [CrossRef] [Green Version]
  81. Syed, Z.A.; Hard, T.; Uv, A.; van Dijk-Hard, I.F. A potential role for Drosophila mucins in development and physiology. PLoS ONE 2008, 3, e3041. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Buchon, N.; Broderick, N.A.; Poidevin, M.; Pradervand, S.; Lemaitre, B. Drosophila intestinal response to bacterial infection: Activation of host defense and stem cell proliferation. Cell Host Microbe 2009, 5, 200–211. [Google Scholar] [CrossRef] [PubMed]
  83. Sauvanet, C.; Wayt, J.; Pelaseyed, T.; Bretscher, A. Structure, regulation, and functional diversity of microvilli on the apical domain of epithelial cells. Annu. Rev. Cell. Dev. Biol. 2015, 31, 593–621. [Google Scholar] [CrossRef]
  84. Polenogova, O.V.; Noskov, Y.A.; Artemchenko, A.S.; Zhangissina, S.; Klementeva, T.N.; Yaroslavtseva, O.N.; Khodyrev, V.P.; Kruykova, N.A.; Glupov, V.V. Citrobacter freundii, a natural associate of the Colorado potato beetle, increases larval susceptibility to Bacillus thuringiensis. Pest Manag. Sci. 2022, 78, 3823–3835. [Google Scholar] [CrossRef] [PubMed]
  85. Hegan, P.S.; Mermall, V.; Tilney, L.G.; Mooseker, M.S. Roles for Drosophila melanogaster myosin IB in maintenance of enterocyte brush-border structure and resistance to the bacterial pathogen Pseudomonas entomophila. Mol. Biol. Cell. 2007, 18, 4625–4636. [Google Scholar] [CrossRef] [Green Version]
  86. Bauer, E.; Lampert, N.; Mikaelyan, A.; Kohler, T.; Maekawa, K.; Brune, A. Physicochemical conditions, metabolites and community structure of the bacterial microbiota in the gut of wood-feeding cockroaches (Blaberidae: Panesthiinae). FEMS Microbiol. Ecol. 2015, 91, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Miao, Z.; Cao, X.; Jiang, H. Digestion-related proteins in the tobacco hornworm, Manduca sexta. Insect Biochem. Mol. Biol. 2020, 126, 103457. [Google Scholar] [CrossRef] [PubMed]
  88. Simser, J.A.; Macaluso, K.R.; Mulenga, A.; Azad, A.F. Immune-responsive lysozymes from hemocytes of the American dog tick, Dermacentor variabilis and an embryonic cell line of the Rocky Mountain wood tick, D. andersoni. Insect Biochem. Mol. Biol. 2004, 34, 1235–1246. [Google Scholar] [CrossRef] [PubMed]
  89. Kim, J.K.; Han, S.H.; Kim, C.H.; Jo, Y.H.; Futahashi, R.; Kikuchi, Y.; Fukatsu, T.; Lee, B.L. Molting-associated suppression of symbiont population and up-regulation of antimicrobial activity in the midgut symbiotic organ of the Riptortus-Burkholderia symbiosis. Dev. Comp. Immunol. 2014, 43, 10–14. [Google Scholar] [CrossRef]
  90. Marra, A.; Hanson, M.A.; Kondo, S.; Erkosar, B.; Lemaitre, B. Drosophila antimicrobial peptides and lysozymes regulate gut microbiota composition and abundance. mBio 2021, 12, e0082421. [Google Scholar] [CrossRef]
  91. Yao, F.; Li, Z.; Zhang, Y.; Zhang, S. A novel short peptidoglycan recognition protein in amphioxus: Identification, expression and bioactivity. Dev. Comp. Immunol. 2012, 38, 332–341. [Google Scholar] [CrossRef] [PubMed]
  92. Duplais, C.; Sarou-Kanian, V.; Massiot, D.; Hassan, A.; Perrone, B.; Estevez, Y.; Wertz, J.T.; Martineau, E.; Farjon, J.; Giraudeau, P.; et al. Gut bacteria are essential for normal cuticle development in herbivorous turtle ants. Nat. Commun. 2021, 12, 676. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, X.; Zhang, F.; Lu, X. Diversity and functional roles of the gut microbiota in Lepidopteran insects. Microorganisms 2022, 10, 1234. [Google Scholar] [CrossRef] [PubMed]
  94. Hassan, B.; Siddiqui, J.A.; Xu, Y. Vertically transmitted gut bacteria and nutrition influence the immunity and fitness of Bactrocera dorsalis larvae. Front. Microbiol. 2020, 11, 596352. [Google Scholar] [CrossRef] [PubMed]
  95. Sansone, C.L.; Cohen, J.; Yasunaga, A.; Xu, J.; Osborn, G.; Subramanian, H.; Gold, B.; Buchon, N.; Cherry, S. Microbiota-dependent priming of antiviral intestinal immunity in Drosophila. Cell Host Microbe. 2015, 18, 571–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Batista, K.K.S.; Vieira, C.S.; Figueiredo, M.B.; Costa-Latge, S.G.; Azambuja, P.; Genta, F.A.; Castro, D.P. Influence of Serratia marcescens and Rhodococcus rhodnii on the Humoral Immunity of Rhodnius prolixus. Int. J. Mol. Sci. 2021, 22, 10901. [Google Scholar] [CrossRef]
  97. Liu, Q.X.; Su, Z.P.; Liu, H.H.; Lu, S.P.; Ma, B.; Zhao, Y.; Hou, Y.M.; Shi, Z.H. The Effect of Gut Bacteria on the Physiology of Red Palm Weevil, Rhynchophorus ferrugineus Olivier and Their Potential for the Control of This Pest. Insects 2021, 12, 594. [Google Scholar] [CrossRef] [PubMed]
  98. Gao, H.; Cui, C.; Wang, L.; Jacobs-Lorena, M.; Wang, S. Mosquito microbiota and implications for disease control. Trends Parasitol. 2020, 36, 98–111. [Google Scholar] [CrossRef]
  99. Zhang, X.; Feng, H.; He, J.; Liang, X.; Zhang, N.; Shao, Y.; Zhang, F.; Lu, X. The gut commensal bacterium Enterococcus faecalis LX10 contributes to defending against Nosema bombycis infection in Bombyx mori. Pest Manag. Sci. 2022, 78, 2215–2227. [Google Scholar] [CrossRef]
  100. Liu, X.; Nagy, P.; Bonfini, A.; Houtz, P.; Bing, X.L.; Yang, X.W.; Buchon, N. Microbes affect gut epithelial cell composition through immune-dependent regulation of intestinal stem cell differentiation. Cell Rep. 2022, 38, 110572. [Google Scholar] [CrossRef]
  101. Muhammad, A.; Habineza, P.; Ji, T.; Hou, Y.; Shi, Z. Intestinal microbiota confer protection by priming the immune system of red palm weevil Rhynchophorus ferrugineus Olivier (Coleoptera: Dryophthoridae). Front. Physiol. 2019, 10, 1303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Hughes, G.L.; Vega-Rodriguez, J.; Xue, P.; Rasgon, J.L. Wolbachia strain wAlbB enhances infection by the rodent malaria parasite Plasmodium berghei in Anopheles gambiae mosquitoes. Appl. Environ. Microbiol. 2012, 78, 1491–1495. [Google Scholar] [CrossRef] [PubMed]
  103. Wei, G.; Lai, Y.L.; Wang, G.D.; Chen, H.; Li, F.; Wang, S.B. Insect pathogenic fungus interacts with the gut microbiota to accelerate mosquito mortality. Proc. Natl. Acad. Sci. USA 2017, 114, 5994–5999. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Neyen, C.; Poidevin, M.; Roussel, A.; Lemaitre, B. Tissue- and ligand-specific sensing of gram-negative infection in Drosophila by PGRP-LC isoforms and PGRP-LE. J. Immunol. 2012, 189, 1886–1897. [Google Scholar] [CrossRef] [Green Version]
  105. Lu, Y.; Su, F.; Li, Q.; Zhang, J.; Li, Y.; Tang, T.; Hu, Q.; Yu, X.Q. Pattern recognition receptors in Drosophila immune responses. Dev. Comp. Immunol. 2020, 102, 103468. [Google Scholar] [CrossRef]
  106. Yu, Y.; Park, J.W.; Kwon, H.M.; Hwang, H.O.; Jang, I.H.; Masuda, A.; Kurokawa, K.; Nakayama, H.; Lee, W.J.; Dohmae, N.; et al. Diversity of innate immune recognition mechanism for bacterial polymeric meso-diaminopimelic acid-type peptidoglycan in insects. J. Biol. Chem. 2010, 285, 32937–32945. [Google Scholar] [CrossRef] [Green Version]
  107. Gendrin, M.; Welchman, D.P.; Poidevin, M.; Herve, M.; Lemaitre, B. Long-range activation of systemic immunity through peptidoglycan diffusion in Drosophila. PLoS Genet. 2009, 5, e1000694. [Google Scholar] [CrossRef] [Green Version]
  108. Iatsenko, I.; Kondo, S.; Mengin-Lecreulx, D.; Lemaitre, B. PGRP-SD, an extracellular pattern-recognition receptor, enhances peptidoglycan-mediated activation of the Drosophila Imd pathway. Immunity 2016, 45, 1013–1023. [Google Scholar] [CrossRef] [Green Version]
  109. Bischoff, V.; Vignal, C.; Duvic, B.; Boneca, I.G.; Hoffmann, J.A.; Royet, J. Downregulation of the Drosophila immune response by peptidoglycan-recognition proteins SC1 and SC2. PLoS Pathog. 2006, 2, e14. [Google Scholar] [CrossRef] [Green Version]
  110. Guo, L.; Karpac, J.; Tran, S.L.; Jasper, H. PGRP-SC2 promotes gut immune homeostasis to limit commensal dysbiosis and extend lifespan. Cell 2014, 156, 109–122. [Google Scholar] [CrossRef] [Green Version]
  111. Aggarwal, K.; Rus, F.; Vriesema-Magnuson, C.; Erturk-Hasdemir, D.; Paquette, N.; Silverman, N. Rudra interrupts receptor signaling complexes to negatively regulate the IMD pathway. PLoS Pathog. 2008, 4, e1000120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kleino, A.; Myllymaki, H.; Kallio, J.; Vanha-aho, L.M.; Oksanen, K.; Ulvila, J.; Hultmark, D.; Valanne, S.; Ramet, M. Pirk is a negative regulator of the Drosophila Imd pathway. J. Immunol. 2008, 180, 5413–5422. [Google Scholar] [CrossRef] [PubMed]
  113. Maillet, F.; Bischoff, V.; Vignal, C.; Hoffmann, J.; Royet, J. The Drosophila peptidoglycan recognition protein PGRP-LF blocks PGRP-LC and IMD/JNK pathway activation. Cell Host Microbe 2008, 3, 293–303. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Cao, Y.; Chtarbanova, S.; Petersen, A.J.; Ganetzky, B. Dnr1 mutations cause neurodegeneration in Drosophila by activating the innate immune response in the brain. Proc. Natl. Acad. Sci. USA 2013, 110, E1752–E1760. [Google Scholar] [CrossRef] [Green Version]
  115. Li, X.M.; Meng, K.; Qiao, J.L.; Liu, H.; Zhong, C.Y.; Liu, Q.Z. Identification of Aadnr1, a novel gene related to innate immunity and apoptosis in Aedes albopictus. Gene 2016, 587, 18–26. [Google Scholar] [CrossRef]
  116. Yokoi, K.; Ito, W.; Kato, D.; Miura, K. RNA interference-based characterization of Caspar, DREDD and FADD genes in immune signaling pathways of the red fl our beetle, Tribolium castaneum (Coleoptera: Tenebrionidae). Eur. J. Entomol. 2022, 119, 23–35. [Google Scholar] [CrossRef]
  117. Garver, L.S.; Dong, Y.; Dimopoulos, M.G. Caspar controls resistance to Plasmodium falciparum in diverse Anopheline species. PLoS Pathog. 2009, 5, e1000335. [Google Scholar] [CrossRef] [Green Version]
  118. Kim, M.; Lee, J.H.; Lee, S.Y.; Kim, E.; Chung, J. Caspar, a suppressor of antibacterial immunity in Drosophila. Proc. Natl. Acad. Sci. USA 2006, 103, 16358–16363. [Google Scholar] [CrossRef] [Green Version]
  119. Fernando, M.D.; Kounatidis, I.; Ligoxygakis, P. Loss of Trabid, a new negative regulator of the drosophila immune-deficiency pathway at the level of TAK1, reduces life span. PLoS Genet. 2014, 10, e1004117. [Google Scholar] [CrossRef] [Green Version]
  120. Aparicio, R.; Neyen, C.; Lemaitre, B.; Busturia, A. dRYBP contributes to the negative regulation of the Drosophila Imd pathway. PLoS ONE 2013, 8, e62052. [Google Scholar] [CrossRef] [Green Version]
  121. Khush, R.S.; Cornwell, W.D.; Uram, J.N.; Lemaitre, B. A ubiquitin-proteasome pathway represses the Drosophila immune deficiency signaling cascade. Curr. Biol. 2002, 12, 1728–1737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Sarvari, M.; Mikani, A.; Mehrabadi, M. The innate immune gene Relish and Caudal jointly contribute to the gut immune homeostasis by regulating antimicrobial peptides in Galleria mellonella. Dev. Comp. Immunol. 2020, 110, 103732. [Google Scholar] [CrossRef] [PubMed]
  123. Cabral, S.; de Paula, A.; Samuels, R.; da Fonseca, R.; Gomes, S.; Silva, J.R.; Mury, F. Aedes aegypti (Diptera: Culicidae) immune responses with different feeding regimes following infection by the entomopathogenic fungus Metarhizium anisopliae. Insects 2020, 11, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Dantoft, W.; Davis, M.M.; Lindvall, J.M.; Tang, X.; Uvell, H.; Junell, A.; Beskow, A.; Engstrom, Y. The Oct1 homolog Nubbin is a repressor of NF-kappaB-dependent immune gene expression that increases the tolerance to gut microbiota. BMC Biol. 2013, 11, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Wehbe, L.S.; Barakat, D.; Acker, A.; El Khoury, R.; Reichhart, J.M.; Matt, N.; El Chamy, L. Protein phosphatase 4 negatively regulates the protein phosphatase 4 negatively regulates the immune deficiency-NF-KB pathway during the Drosophila immune response. J. Immunol. 2021, 207, 1616–1626. [Google Scholar] [CrossRef] [PubMed]
  126. Li, R.M.; Zhou, H.J.; Jia, C.L.; 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. [Google Scholar] [CrossRef]
  127. Ha, E.M.; Oh, C.T.; Ryu, J.H.; Bae, Y.S.; Kang, S.W.; Jang, I.H.; Brey, P.T.; Lee, W.J. An antioxidant system required for host protection against gut infection in Drosophila. Dev. Cell. 2005, 8, 125–132. [Google Scholar] [CrossRef] [Green Version]
  128. Yao, Z.; Wang, A.; Li, Y.; Cai, Z.; Lemaitre, B.; Zhang, H. The dual oxidase gene BdDuox regulates the intestinal bacterial community homeostasis of Bactrocera dorsalis. ISME J. 2016, 10, 1037–1050. [Google Scholar] [CrossRef] [Green Version]
  129. Zeng, T.; Su, H.A.; Liu, Y.L.; Li, J.F.; Jiang, D.X.; Lu, Y.Y.; Qi, Y.X. Serotonin modulates insect gut bacterial community homeostasis. BMC Biol. 2022, 20, 105. [Google Scholar] [CrossRef]
  130. Xiao, X.; Yang, L.; Pang, X.; Zhang, R.; Zhu, Y.; Wang, P.; Gao, G.; Cheng, G. A Mesh-Duox pathway regulates homeostasis in the insect gut. Nat. Microbiol. 2017, 2, 17020. [Google Scholar] [CrossRef] [Green Version]
  131. Chakrabarti, S.; Poidevin, M.; Lemaitre, B. The Drosophila MAPK p38c regulates oxidative stress and lipid homeostasis in the intestine. PLoS Genet. 2014, 10, e1004659. [Google Scholar] [CrossRef] [PubMed]
  132. Lee, K.A.; Kim, S.H.; Kim, E.K.; Ha, E.M.; You, H.; Kim, B.; Kim, M.J.; Kwon, Y.; Ryu, J.H.; Lee, W.J. Bacterial-derived uracil as a modulator of mucosal immunity and gut-microbe homeostasis in Drosophila. Cell 2013, 153, 797–811. [Google Scholar] [CrossRef] [PubMed]
  133. Lee, K.A.; Kim, B.; Bhin, J.; Kim, D.H.; You, H.; Kim, E.K.; Kim, S.H.; Ryu, J.H.; Hwang, D.; Lee, W.J. Bacterial uracil modulates Drosophila DUOX-dependent gut immunity via Hedgehog-induced signaling endosomes. Cell Host Microbe 2015, 17, 191–204. [Google Scholar] [CrossRef] [Green Version]
  134. Ha, E.M.; Lee, K.A.; Park, S.H.; Kim, S.H.; Nam, H.J.; Lee, H.Y.; Kang, D.; Lee, W.J. Regulation of DUOX by the Galphaq-phospholipase Cbeta-Ca2+ pathway in Drosophila gut immunity. Dev. Cell. 2009, 16, 386–397. [Google Scholar] [CrossRef] [Green Version]
  135. Ha, E.M.; Lee, K.A.; Seo, Y.Y.; Kim, S.H.; Lim, J.H.; Oh, B.H.; Kim, J.; Lee, W.J. Coordination of multiple dual oxidase-regulatory pathways in responses to commensal and infectious microbes in Drosophila gut. Nat. Immunol. 2009, 10, 949–957. [Google Scholar] [CrossRef]
  136. Hochmuth, C.E.; Biteau, B.; Bohmann, D.; Jasper, H. Redox regulation by Keap1 and Nrf2 controls intestinal stem cell proliferation in Drosophila. Cell Stem Cell 2011, 8, 188–199. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Pitoniak, A.; Bohmann, D. Mechanisms and functions of Nrf2 signaling in Drosophila. Free Radic. Biol. Med. 2015, 88, 302–313. [Google Scholar] [CrossRef] [Green Version]
  138. Chakrabarti, S.; Dudzic, J.P.; Li, X.; Collas, E.J.; Boquete, J.P.; Lemaitre, B. Remote control of intestinal Stem Cell activity by haemocytes in Drosophila. PLoS Genet. 2016, 12, e1006089. [Google Scholar] [CrossRef] [Green Version]
  139. Malagoli, D.; Sacchi, S.; Ottaviani, E. Unpaired (upd)-3 expression and other immune-related functions are stimulated by interleukin-8 in Drosophila melanogaster SL2 cell line. Cytokine 2008, 44, 269–274. [Google Scholar] [CrossRef]
  140. Yu, S.; Luo, F.; Xu, Y.; Zhang, Y.; Jin, L.H. Drosophila innate immunity involves multiple signaling pathways and coordinated communication between different tissues. Front. Immunol. 2022, 13, 905370. [Google Scholar] [CrossRef]
  141. Myllymaki, H.; Ramet, M. JAK/STAT pathway in Drosophila immunity. Scand. J. Immunol. 2014, 79, 377–385. [Google Scholar] [CrossRef] [PubMed]
  142. Bang, I.S. JAK/STAT signaling in insect innate immunity. Entomol. Res. 2019, 49, 339–353. [Google Scholar] [CrossRef]
  143. West, C.; Silverman, N. p38b and JAK-STAT signaling protect against Invertebrate iridescent virus 6 infection in Drosophila. PLoS Pathog. 2018, 14, e1007020. [Google Scholar] [CrossRef]
  144. Ahlers, L.R.H.; Trammell, C.E.; Carrell, G.F.; Mackinnon, S.; Torrevillas, B.K.; Chow, C.Y.; Luckhart, S.; Goodman, A.G. Insulin potentiates JAK/STAT signaling to broadly inhibit flavivirus replication in insect vectors. Cell Rep. 2019, 29, 1946–1960.e5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Trammell, C.E.; Goodman, A.G. Emerging mechanisms of insulin-mediated antiviral immunity in Drosophila melanogaster. Front. Immunol. 2019, 10, 2973. [Google Scholar] [CrossRef] [PubMed]
  146. Jiang, L. Insights into the antiviral pathways of the Silkworm Bombyx mori. Front. Immunol. 2021, 12, 639092. [Google Scholar] [CrossRef] [PubMed]
  147. Ma, M.; Guo, L.; Tu, C.; Wang, A.; Xu, L.; Luo, J. Comparative analysis of Adelphocoris suturalis Jakovlev (Hemiptera: Miridae) immune responses to fungal and bacterial pathogens. Front. Physiol. 2021, 12, 646721. [Google Scholar] [CrossRef]
  148. Geng, T.; Lu, F.; Wu, H.; Lou, D.; Tu, N.; Zhu, F.; Wang, S. Target antifungal peptides of immune signalling pathways in silkworm, Bombyx mori, against Beauveria bassiana. Insect Mol. Biol. 2021, 30, 102–112. [Google Scholar] [CrossRef]
  149. Lokau, J.; Schoeder, V.; Haybaeck, J.; Garbers, C. Jak-Stat signaling Induced by interleukin-6 family cytokines in Hepatocellular Carcinoma. Cancers 2019, 11, 1704. [Google Scholar] [CrossRef] [Green Version]
  150. Owen, K.L.; Brockwell, N.K.; Parker, B.S. JAK-STAT signaling: A double-edged sword of immune regulation and cancer progression. Cancers 2019, 11, 2002. [Google Scholar] [CrossRef] [Green Version]
  151. Ghiglione, C.; Devergne, O.; Georgenthum, E.; Carballes, F.; Medioni, C.; Cerezo, D.; Noselli, S. The Drosophila cytokine receptor Domeless controls border cell migration and epithelial polarization during oogenesis. Development 2002, 129, 5437–5447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Moore, R.; Vogt, K.; Acosta-Martin, A.E.; Shire, P.; Zeidler, M.; Smythe, E. Integration of JAK/STAT receptor-ligand trafficking, signalling and gene expression in Drosophila melanogaster cells. J. Cell Sci. 2020, 133, jcs246199. [Google Scholar] [CrossRef] [PubMed]
  153. Luo, H.; Rose, P.E.; Roberts, T.M.; Dearolf, C.R. The Hopscotch Jak kinase requires the Raf pathway to promote blood cell activation and differentiation in Drosophila. Mol. Genet. Genom. 2002, 267, 57–63. [Google Scholar] [CrossRef] [PubMed]
  154. Hao, Y.; Pan, J.; Chen, Q.; Gu, H.; Ji, G.; Yue, G.; Yang, S. Jumu is required for the activation of JAK/STAT in Drosophila lymph gland development and epidermal wounds. Biochem. Biophys. Res. Commun. 2022, 591, 68–75. [Google Scholar] [CrossRef]
  155. Gronholm, J.; Ungureanu, D.; Vanhatupa, S.; Ramet, M.; Silvennoinen, O. Sumoylation of Drosophila transcription factor STAT92E. J. Innate Immun. 2010, 2, 618–624. [Google Scholar] [CrossRef]
  156. Vollmer, J.; Fried, P.; Aguilar-Hidalgo, D.; Sanchez-Aragon, M.; Iannini, A.; Casares, F.; Iber, D. Growth control in the Drosophila eye disc by the cytokine Unpaired. Development 2017, 144, 837–843. [Google Scholar] [CrossRef] [Green Version]
  157. Okugawa, S.; Mekata, T.; Inada, M.; Kihara, K.; Shiki, A.; Kannabiran, K.; Kono, T.; Sakai, M.; Yoshida, T.; Itami, T.; et al. The SOCS and STAT from JAK/STAT signaling pathway of kuruma shrimp Marsupenaeus japonicus: Molecular cloning, characterization and expression analysis. Mol. Cell. Probes 2013, 27, 6–14. [Google Scholar] [CrossRef]
  158. Stec, W.; Vidal, O.; Zeidler, M.P. Drosophila SOCS36E negatively regulates JAK/STAT pathway signaling via two separable mechanisms. Mol. Biol. Cell 2013, 24, 3000–3009. [Google Scholar] [CrossRef]
  159. Monahan, A.J.; Starz-Gaiano, M. Socs36E limits STAT signaling via Cullin2 and a SOCS-box independent mechanism in the Drosophila egg chamber. Mech. Dev. 2015, 138, 313–327. [Google Scholar] [CrossRef]
  160. Ren, W.; Zhang, Y.; Li, M.; Wu, L.; Wang, G.; Baeg, G.H.; You, J.; Li, Z.; Lin, X. Windpipe controls Drosophila intestinal homeostasis by regulating JAK/STAT pathway via promoting receptor endocytosis and lysosomal degradation. PLoS Genet. 2015, 11, e1005180. [Google Scholar] [CrossRef]
  161. Barillas-Mury, C.; Han, Y.S.; Seeley, D.; Kafatos, F.C. Anopheles gambiae Ag-STAT, a new insect member of the STAT family, is activated in response to bacterial infection. EMBO J. 1999, 18, 959–967. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Osman, D.; Buchon, N.; Chakrabarti, S.; Huang, Y.T.; Su, W.C.; Poidevin, M.; Tsai, Y.C.; Lemaitre, B. Autocrine and paracrine unpaired signaling regulate intestinal stem cell maintenance and division. J. Cell Sci. 2012, 125, 5944–5949. [Google Scholar] [CrossRef] [PubMed]
  163. Bonfini, A.; Liu, X.; Buchon, N. From pathogens to microbiota: How Drosophila intestinal stem cells react to gut microbes. Dev. Comp. Immunol. 2016, 64, 22–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Buchon, N.; Broderick, N.A.; Chakrabarti, S.; Lemaitre, B. Invasive and indigenous microbiota impact intestinal stem cell activity through multiple pathways in Drosophila. Gene Dev. 2009, 23, 2333–2344. [Google Scholar] [CrossRef] [Green Version]
  165. Nan, Y.C.; Wu, C.Y.; Zhang, Y.J. Interplay between janus kinase/signal transducer and activator of transcription signaling activated by type I interferons and viral antagonism. Front. Immunol. 2017, 8, 1758. [Google Scholar] [CrossRef]
  166. Ezeonwumelu, I.J.; Garcia-Vidal, E.; Ballana, E. JAK-STAT pathway: A novel target to tackle viral infections. Viruses 2021, 13, 2379. [Google Scholar] [CrossRef]
  167. Cerenius, L.; Kawabata, S.; Lee, B.L.; Nonaka, M.; Soderhall, K. Proteolytic cascades and their involvement in invertebrate immunity. Trends Biochem. Sci. 2010, 35, 575–583. [Google Scholar] [CrossRef]
  168. Barillas-Mury, C. CLIP proteases and Plasmodium melanization in Anopheles gambiae. Trends Parasitol. 2007, 23, 297–299. [Google Scholar] [CrossRef]
  169. Sugumaran, M. Comparative biochemistry of eumelanogenesis and the protective roles of phenoloxidase and melanin in insects. Pigment Cell Res. 2002, 15, 2–9. [Google Scholar] [CrossRef]
  170. Charoensapsri, W.; Amparyup, P.; Suriyachan, C.; Tassanakajon, A. Melanization reaction products of shrimp display antimicrobial properties against their major bacterial and fungal pathogens. Dev. Comp. Immunol. 2014, 47, 150–159. [Google Scholar] [CrossRef] [Green Version]
  171. Blandin, S.A.; Marois, E.; Levashina, E.A. Antimalarial responses in Anopheles gambiae: From a complement-like protein to a complement-like pathway. Cell Host Microbe 2008, 3, 364–374. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Whitten, M.M.; Shiao, S.H.; Levashina, E.A. Mosquito midguts and malaria: Cell biology, compartmentalization and immunology. Parasite Immunol. 2006, 28, 121–130. [Google Scholar] [CrossRef] [PubMed]
  173. Christophides, G.K.; Vlachou, D.; Kafatos, F.C. Comparative and functional genomics of the innate immune system in the malaria vector Anopheles gambiae. Immunol. Rev. 2004, 198, 127–148. [Google Scholar] [CrossRef] [PubMed]
  174. Shao, Q.; Yang, B.; Xu, Q.; Li, X.; Lu, Z.; Wang, C.; Huang, Y.; Soderhall, K.; Ling, E. Hindgut innate immunity and regulation of fecal microbiota through melanization in insects. J. Biol. Chem. 2012, 287, 14270–14279. [Google Scholar] [CrossRef] [Green Version]
  175. Wu, K.; Wang, J.; Geng, L.; Chen, K.; Huang, W.; Liu, Q.; Beerntsen, B.T.; Ling, E. Loss of control of the culturable bacteria in the hindgut of Bombyx mori after Cry1Ab ingestion. Dev. Comp. Immunol. 2020, 111, 103754. [Google Scholar] [CrossRef]
  176. Yuan, C.; Xing, L.; Wang, M.; Hu, Z.; Zou, Z. Microbiota modulates gut immunity and promotes baculovirus infection in Helicoverpa armigera. Insect Sci. 2021, 28, 1766–1779. [Google Scholar] [CrossRef]
  177. Didier, E.S.; Didier, P.J.; Snowden, K.F.; Shadduck, J.A. Microsporidiosis in mammals. Microbes Infect. 2000, 2, 709–720. [Google Scholar] [CrossRef]
  178. Ma, Z.; Li, C.; Pan, G.; Li, Z.; Han, B.; Xu, J.; Lan, X.; Chen, J.; Yang, D.; Chen, Q.; et al. Genome-wide transcriptional response of silkworm (Bombyx mori) to infection by the microsporidian Nosema bombycis. PLoS ONE 2013, 8, e84137. [Google Scholar] [CrossRef] [Green Version]
  179. Gegear, R.J.; Otterstatter, M.C.; Thomson, J.D. Bumble-bee foragers infected by a gut parasite have an impaired ability to utilize floral information. Proc. R. Soc. B Biol. Sci. 2006, 273, 1073–1078. [Google Scholar] [CrossRef] [Green Version]
  180. Chen, Q.; Wei, T. Viral receptors of the gut: Insect-borne propagative plant viruses of agricultural importance. Curr. Opin. Insect Sci. 2016, 16, 9–13. [Google Scholar] [CrossRef] [Green Version]
  181. Ponnuvel, K.M.; Nakazawa, H.; Furukawa, S.; Asaoka, A.; Ishibashi, J.; Tanaka, H.; Yamakawa, M. A lipase isolated from the silkworm Bombyx mori shows antiviral activity against nucleopolyhedrovirus. J. Virol. 2003, 77, 10725–10729. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Ponnuvel, K.M.; Nithya, K.; Sirigineedi, S.; Awasthi, A.K.; Yamakawa, M. In Vitro antiviral activity of an alkaline trypsin from the digestive juice of Bombyx mori larvae against nucleopolyhedrovirus. Arch. Insect. Biochem. Physiol. 2012, 81, 90–104. [Google Scholar] [CrossRef]
  183. Wu, P.; Wang, X.; Qin, G.X.; Liu, T.; Jiang, Y.F.; Li, M.W.; Guo, X.J. Microarray analysis of the gene expression profile in the midgut of silkworm infected with cytoplasmic polyhedrosis virus. Mol. Biol. Rep. 2011, 38, 333–341. [Google Scholar] [CrossRef] [PubMed]
  184. Clem, R.J. The role of apoptosis in defense against baculovirus infection in insects. Curr. Top. Microbiol. Immunol. 2005, 289, 113–129. [Google Scholar] [PubMed]
  185. Zhang, P.; Edgar, B.A. Insect gut regeneration. Cold Spring Harb. Perspect. Biol. 2022, 14, a040915. [Google Scholar] [CrossRef] [PubMed]
  186. Biteau, B.; Hochmuth, C.E.; Jasper, H. JNK activity in somatic stem cells causes loss of tissue homeostasis in the aging Drosophila gut. Cell Stem Cell. 2008, 3, 442–455. [Google Scholar] [CrossRef] [Green Version]
  187. Cordero, J.B.; Stefanatos, R.K.; Scopelliti, A.; Vidal, M.; Sansom, O.J. Inducible progenitor-derived Wingless regulates adult midgut regeneration in Drosophila. EMBO J. 2012, 31, 3901–3917. [Google Scholar] [CrossRef] [Green Version]
  188. Karpowicz, P.; Perez, J.; Perrimon, N. The Hippo tumor suppressor pathway regulates intestinal stem cell regeneration. Development 2010, 137, 4135–4145. [Google Scholar] [CrossRef] [Green Version]
  189. Neophytou, C.; Pitsouli, C. How gut microbes nurture intestinal Stem Cells: A Drosophila perspective. Metabolites 2022, 12, 169. [Google Scholar] [CrossRef]
  190. Yadav, S.; Eleftherianos, I. Participation of the Serine Protease Jonah66Ci in the Drosophila antinematode immune response. Infect. Immun. 2019, 87, e00094-19. [Google Scholar] [CrossRef] [Green Version]
  191. Tang, X.; Engstrom, Y. Regulation of immune and tissue homeostasis by Drosophila POU factors. Insect Biochem. Mol. Biol. 2019, 109, 24–30. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The insect intestinal immune defense system. The insect intestinal immune defense system consists of a physical barrier and an immune response. Pathogens entering the intestinal epithelium face a physical barrier such as the mucus layer of the peritrophic membrane and intestinal cells. A number of signaling molecules produced by pathogens activate a range of immune responses: the Imd signaling pathway and the JAK/STAT signaling pathway produce AMPs, Duox produces ROS, and the melanization response produces a range of bactericidal active substances. This whole system effectively regulates the balance of gut microbes. PM: peritrophic membrane; AMPs, Antimicrobial peptides; ROS, reactive oxygen species; PPO, polyphenol oxidase.
Figure 1. The insect intestinal immune defense system. The insect intestinal immune defense system consists of a physical barrier and an immune response. Pathogens entering the intestinal epithelium face a physical barrier such as the mucus layer of the peritrophic membrane and intestinal cells. A number of signaling molecules produced by pathogens activate a range of immune responses: the Imd signaling pathway and the JAK/STAT signaling pathway produce AMPs, Duox produces ROS, and the melanization response produces a range of bactericidal active substances. This whole system effectively regulates the balance of gut microbes. PM: peritrophic membrane; AMPs, Antimicrobial peptides; ROS, reactive oxygen species; PPO, polyphenol oxidase.
Ijms 23 15132 g001
Figure 2. Imd signaling pathway and its negative regulator in insect intestinal immunity (modified from the reference [59]). When pathogens invade the gut, diaminopyrimidine-type peptidoglycan (DAP-PGN) in the cell wall is recognized by the peptidoglycan recognition receptor LC (PGRP-LC) in the cell membrane, activating the intranuclear transcription factor Relish to regulate antimicrobial peptides (AMPs); the intramembrane receptor PGRP-LE recognizes tracheal cytotoxin (TCT) and activates Imd pathway; Pirk is a repressor of PGRP-LE and PGRP-LC activity, PGRP-LF blocks PGRP-LC activity, and Relish transcription is regulated by negative feedback from Caudal genes. tAk1, transforming growth factor-β-activated kinase 1; IKK, inhibitor of kappa B kinase.
Figure 2. Imd signaling pathway and its negative regulator in insect intestinal immunity (modified from the reference [59]). When pathogens invade the gut, diaminopyrimidine-type peptidoglycan (DAP-PGN) in the cell wall is recognized by the peptidoglycan recognition receptor LC (PGRP-LC) in the cell membrane, activating the intranuclear transcription factor Relish to regulate antimicrobial peptides (AMPs); the intramembrane receptor PGRP-LE recognizes tracheal cytotoxin (TCT) and activates Imd pathway; Pirk is a repressor of PGRP-LE and PGRP-LC activity, PGRP-LF blocks PGRP-LC activity, and Relish transcription is regulated by negative feedback from Caudal genes. tAk1, transforming growth factor-β-activated kinase 1; IKK, inhibitor of kappa B kinase.
Ijms 23 15132 g002
Figure 3. Regulation of ROS and the physiological roles of JAK/STAT signaling pathway in insect gut immunity. (A) Regulation of ROS (modified from the reference [59]): When pathogens invade the insect gut, secreted uracil stimulates the formation of Cad99C/PLCβ/PKC upstream signaling endosomes via GPCRs, which releases signaling molecules to regulate the flow of Ca2+ in the endoplasmic reticulum to activate the enzymatic activity of Duox in the cell membrane; Peptidoglycan also regulates the formation of Cad99C through the Hedgehog (Hh) signaling pathway to interact with in Duox enzyme. The transcriptional regulation of the Duox gene is regulated by the intranuclear transcription factor Atf2Atf2, which is regulated by the MEKK1/MEK3/p38 MAPK cascade; at the same time, the signal endosome activated by uracil activates the transcription of ATF2 through PLCβ, and the IMD signaling pathway also activates this pathway; MAKP phosphatase 3 (Mkp3) downregulates Duox expression, and PLCβ and calcium regulate neurophosphatase B (CanB) induce Mkp3 transcription;; Excess ROS in the intestine activates the JAK/STAT and JNK pathways, which transmit signals to the nucleus, enabling the expression of cellular repair signaling factor UPD that initiate repair of the intestine and thus participate in the intestinal immune defense response. (B) The physiological roles of JAK/STAT signaling pathway in insect gut immunity: When pathogens infect the intestine, upd3 ligands are produced in infected cells, which act as signaling molecules into uninfected healthy cells, activating Dro3 expression and intestinal repair. Upd3, the members of the Unpaired family; Dro3: Drosomycin 3.
Figure 3. Regulation of ROS and the physiological roles of JAK/STAT signaling pathway in insect gut immunity. (A) Regulation of ROS (modified from the reference [59]): When pathogens invade the insect gut, secreted uracil stimulates the formation of Cad99C/PLCβ/PKC upstream signaling endosomes via GPCRs, which releases signaling molecules to regulate the flow of Ca2+ in the endoplasmic reticulum to activate the enzymatic activity of Duox in the cell membrane; Peptidoglycan also regulates the formation of Cad99C through the Hedgehog (Hh) signaling pathway to interact with in Duox enzyme. The transcriptional regulation of the Duox gene is regulated by the intranuclear transcription factor Atf2Atf2, which is regulated by the MEKK1/MEK3/p38 MAPK cascade; at the same time, the signal endosome activated by uracil activates the transcription of ATF2 through PLCβ, and the IMD signaling pathway also activates this pathway; MAKP phosphatase 3 (Mkp3) downregulates Duox expression, and PLCβ and calcium regulate neurophosphatase B (CanB) induce Mkp3 transcription;; Excess ROS in the intestine activates the JAK/STAT and JNK pathways, which transmit signals to the nucleus, enabling the expression of cellular repair signaling factor UPD that initiate repair of the intestine and thus participate in the intestinal immune defense response. (B) The physiological roles of JAK/STAT signaling pathway in insect gut immunity: When pathogens infect the intestine, upd3 ligands are produced in infected cells, which act as signaling molecules into uninfected healthy cells, activating Dro3 expression and intestinal repair. Upd3, the members of the Unpaired family; Dro3: Drosomycin 3.
Ijms 23 15132 g003
Table 1. Physical barriers and key gut immunity response in different insects.
Table 1. Physical barriers and key gut immunity response in different insects.
SpeciesPhysical BarriersImmunity Response
Diptera:
Drosophila melanogasterPM [1]
Mucus [2]
Imd [3]
Toll [4]
Duox-ROS [5]
JAK/STAT [6]
NOX-ROS [7]
Melanization [8]
Bactrocera dorsalisPM [9]Imd [10,11]
Duox-ROS [12]
JAK/STAT [9]
Anopheles gambiaePM [13,14]Imd [15]
Duox-ROS [16]
JAK/STAT [17]
Lepidoptera:
Bombyx moriPM [18]Imd [19]
Duox-ROS [20]
JAK/STAT [20]
Plutella xylostellaPM [21]Imd [22]
Toll [22]
JAK/STAT [22]
Hemiptera:
Acyrthosiphon pisumPMM [23]JAK/STAT [24]
Nilaparvata lugensPMM [25]Imd [26]
JAK/STAT [26]
Coleoptera:
Holotrichia oblitaPM [27]_
Rhynchophorus ferrugineus_ _Imd [28]
Toll [29]
JAK/STAT [30]
Hymenoptera:
Apis melliferaPM [31]Imd [32]
Duox-ROS [33]
JAK/STAT [34]
Melanization [35]
Bombus muscorum_ _Melanization [36]
“_” indicates that no studies on intestinal immunity have been reported in this species; “_ _” indicates that such species has not been reported to have PM or mucus.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Zeng, T.; Jaffar, S.; Xu, Y.; Qi, Y. The Intestinal Immune Defense System in Insects. Int. J. Mol. Sci. 2022, 23, 15132. https://doi.org/10.3390/ijms232315132

AMA Style

Zeng T, Jaffar S, Xu Y, Qi Y. The Intestinal Immune Defense System in Insects. International Journal of Molecular Sciences. 2022; 23(23):15132. https://doi.org/10.3390/ijms232315132

Chicago/Turabian Style

Zeng, Tian, Saleem Jaffar, Yijuan Xu, and Yixiang Qi. 2022. "The Intestinal Immune Defense System in Insects" International Journal of Molecular Sciences 23, no. 23: 15132. https://doi.org/10.3390/ijms232315132

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop